1
|
Lan X, Johnston E, Ning T, Chen G, Haglund L, Li J. Immunomodulatory bioadhesive technologies. Biomaterials 2025; 321:123274. [PMID: 40156979 DOI: 10.1016/j.biomaterials.2025.123274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Bioadhesives have found significant use in medicine and engineering, particularly for wound care, tissue engineering, and surgical applications. Compared to traditional wound closure methods such as sutures and staples, bioadhesives offer advantages, including reduced tissue damage, enhanced healing, and ease of implementation. Recent progress highlights the synergy of bioadhesives and immunoengineering strategies, leading to immunomodulatory bioadhesives capable of modulating immune responses at local sites where bioadhesives are applied. They foster favorable therapeutic outcomes such as reduced inflammation in wounds and implants or enhanced local immune responses to improve cancer therapy efficacy. The dual functionalities of bioadhesion and immunomodulation benefit wound management, tissue regeneration, implantable medical devices, and post-surgical cancer management. This review delves into the interplay between bioadhesion and immunomodulation, highlighting the mechanobiological coupling involved. Key areas of focus include the modulation of immune responses through chemical and physical strategies, as well as the application of these bioadhesives in wound healing and cancer treatment. Discussed are remaining challenges such as achieving long-term stability and effectiveness, necessitating further research to fully harness the clinical potential of immunomodulatory bioadhesives.
Collapse
Affiliation(s)
- Xiaoyi Lan
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada
| | - Evan Johnston
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada
| | - Tianqin Ning
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada; Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada; Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Pine Ave W, Montreal, Quebec, H3A 1A3, Canada
| | - Lisbet Haglund
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Shriners Hospital for Children, 1003 Decarie Blvd, Montreal, Quebec, H4A 0A9, Canada.
| | - Jianyu Li
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada; Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada.
| |
Collapse
|
2
|
Bi X, Mao Z, Zhang Y, Ren Z, Yang K, Yu C, Chen L, Zheng R, Guan J, Liu Z, Yu B, Huang Y, Shu X, Zheng Y. Endogenous dual-responsive and self-adaptive silk fibroin-based scaffold with enhancement of immunomodulation for skull regeneration. Biomaterials 2025; 320:123261. [PMID: 40132357 DOI: 10.1016/j.biomaterials.2025.123261] [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: 12/15/2024] [Revised: 02/19/2025] [Accepted: 03/16/2025] [Indexed: 03/27/2025]
Abstract
Despite the current biomaterials (e.g. titanium mesh and polyether ether ketone) have been applied to clinical skull repair, the limitations on mechanical match, shape adaptability, bioactivity and osteointegration have greatly limited their clinical application. In this work, we constructed a water and inflammatory microenvironment dual-responsive self-adaptive silk fibroin-magnesium oxide-based scaffold with the matrix metalloproteinase-2-responsive gelatin-methacryloyl-interleukin-4 (IL-4) coating, which presented good mechanical compliance, quickly shape matching and intraoperative reprocessability. With the capability of responding to an acute inflammation microenvironment followed by a triggered on-demand release of the IL-4, the combination of immunoactive IL-4 and Mg2+ co-ordinately facilitated metabolic reprogramming by suppressing glycolysis, promoting mitochondrial oxidative phosphorylation and modulating adenosine 5'-monophosphate-activated protein kinase (AMPK) signalling pathways in macrophages, resulting in significantly facilitating M2 macrophage activation. During the stage of tissue remodelling, the sustained release of Mg2+ further promoted macrophage M2 polarization and the expression of anti-inflammatory cytokines, significantly reduced immune response and improved ectopic osteogenesis ability. Meanwhile, the cranial defect models of male rats demonstrated that this scaffold could significantly enhance biomineralized deposition and vascularisation, and achieve good bone regeneration of cranial defects. Overall, the bioactive scaffold provides a promising biomaterial and alternative repair strategy for critical-size skull defect repair.
Collapse
Affiliation(s)
- Xuewei Bi
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China; School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Zhinan Mao
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China; School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Yilin Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Zeqi Ren
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Kang Yang
- School of Materials Science and Engineering, Anhui University of Technology, Maanshan 243002, China
| | - Chunhao Yu
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China; School of Life, Beijing Institute of Technology, No.5, Zhongguancun South Street, Haidian District, Beijing, China
| | - Lei Chen
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Rui Zheng
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Juan Guan
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science & Engineering, Beihang University, Beijing 100191, China
| | - Zhenhai Liu
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Binsheng Yu
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China
| | - Yongcan Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China.
| | - Xiong Shu
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| |
Collapse
|
3
|
Kabil A, Nayyar N, Xu C, Brassard J, Hill LA, Shin SB, Chopra S, Lo B, Li Y, Bal M, Theret M, Rossi FM, Underhill TM, Hughes MR, McNagny KM. Functional targeting of ILC2s and ILC3s reveals selective roles in intestinal fibrosis and homeostasis. J Exp Med 2025; 222:e20241671. [PMID: 40434419 PMCID: PMC12118370 DOI: 10.1084/jem.20241671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 03/06/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Innate lymphoid cells (ILCs) are long-lived, tissue-resident cell analogs to T helper subsets that lack antigen-specific receptors. Understanding the roles of specific ILCs in chronic inflammation and fibrosis has been limited by inadequate tools for selective targeting. Here, we used Il17rb-CreERT2-eGFP and Rorc-Cre strains to selectively delete RORα in ILC2s and ILC3/Th17 cells, respectively. RORα deletion in ILC2s caused significant loss of gastrointestinal ILC2s, increased ILC3 abundance, elevated Th17-type responses, and heightened susceptibility to Crohn's disease-like fibrosis. Conversely, RORα deletion in ILC3/Th17 cells reduced IL-17 production, protecting against fibrosis. Using isolithocholic acid (isoLCA), a microbial secondary bile acid and RORγt inverse agonist, we confirmed the role of ILC3s/Th17 cells in fibrosis. In RORγt reporter and Th17-deficient Rag1-/- mice, isoLCA reduced IL-17 production by ILC3s and attenuated intestinal fibrosis by dampening RORγt-dependent ILC3/Th17 responses. These findings reveal a novel interplay between ILC2s and ILC3s in gut homeostasis and demonstrate the therapeutic potential of targeting RORγt in ILC3s as a strategy for preventing fibrosis.
Collapse
MESH Headings
- Animals
- Homeostasis/immunology
- Fibrosis
- Mice
- Immunity, Innate
- Lymphocytes/immunology
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Intestines/pathology
- Intestines/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 1/genetics
- Mice, Knockout
- Interleukin-17/metabolism
- Mice, Inbred C57BL
- Crohn Disease/immunology
- Crohn Disease/pathology
Collapse
Affiliation(s)
- Ahmed Kabil
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Natalia Nayyar
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Chengxi Xu
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Julyanne Brassard
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Lesley A. Hill
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Samuel B. Shin
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Sameeksha Chopra
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Bernard Lo
- Department of Pathology and Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Yicong Li
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Mya Bal
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Marine Theret
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Fabio M.V. Rossi
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
- The Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - T. Michael Underhill
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Michael R. Hughes
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Kelly M. McNagny
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
- The Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- The Center for Heart Lung Innovation, St Paul’s Hospital, Vancouver, Canada
| |
Collapse
|
4
|
Guerra P, Ruvoletto M, Quarta S, Boninsegna G, Biasiolo A, Cagnin S, Angeli P, Pontisso P, Martini A. The impact of serpinB3-PD polymorphism on the prognosis of patients with hepatocellular carcinoma. Transl Oncol 2025; 57:102413. [PMID: 40367592 DOI: 10.1016/j.tranon.2025.102413] [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: 07/20/2024] [Revised: 04/17/2025] [Accepted: 05/10/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND HCC ranks as the third leading cause of cancer-related death, yet current surveillance strategies miss over one-third of cases at an early stage. SerpinB3-PD (SB3-PD), a polymorphic isoform of a serine-protease inhibitor involved in tumorigenesis and fibrogenesis, has been related to a more rapid cirrhosis decompensation. This study investigates the prognostic role of SB3-PD in patients with HCC. METHODS SB3-PD polymorphism was assessed in 140 patients with HCC, followed up in our outpatient Clinic. Cell invasion analysis was conducted on HepG2 cells either overexpressing the SB3 wild-type (HepG2/SB3WT) or the PD isoform (HepG2/SB3PD). The effect of recombinant SB3-WT or SB3-PD on the production of molecules that impair immunosurveillance was also assessed in the THP-1 monocytic cell line. RESULTS Patients carrying SB3-PD polymorphism showed worse tumour characteristics, associated with significantly lower survival and SB3-PD was an independent predictor of mortality. HepG2/SB3PD cells had a significantly higher invasion capacity than the HepG2/SB3WT. In THP-1 cells recombinant SB3-PD induced higher levels of PDL1 and IL-13 than SB3-WT. CONCLUSION SB3-PD isoform is associated with worse clinical prognosis in patients with HCC. These findings were supported in vitro by increased cellular invasion and higher production of molecules impairing immunosurveillance.
Collapse
Affiliation(s)
- Pietro Guerra
- Unit of Internal Medicine and Hepatology, Department of Medicine, University of Padova, via Giustiniani, 2, 35128, Padova, Italy
| | - Mariagrazia Ruvoletto
- Unit of Internal Medicine and Hepatology, Department of Medicine, University of Padova, via Giustiniani, 2, 35128, Padova, Italy; European Reference Network - ERN RARELIVER, Department of Medicine, Azienda Ospedale-Università, Padova, Italy
| | - Santina Quarta
- Unit of Internal Medicine and Hepatology, Department of Medicine, University of Padova, via Giustiniani, 2, 35128, Padova, Italy; European Reference Network - ERN RARELIVER, Department of Medicine, Azienda Ospedale-Università, Padova, Italy
| | - Giulia Boninsegna
- Unit of Internal Medicine and Hepatology, Department of Medicine, University of Padova, via Giustiniani, 2, 35128, Padova, Italy
| | - Alessandra Biasiolo
- Unit of Internal Medicine and Hepatology, Department of Medicine, University of Padova, via Giustiniani, 2, 35128, Padova, Italy; European Reference Network - ERN RARELIVER, Department of Medicine, Azienda Ospedale-Università, Padova, Italy
| | - Silvia Cagnin
- Unit of Internal Medicine and Hepatology, Department of Medicine, University of Padova, via Giustiniani, 2, 35128, Padova, Italy
| | - Paolo Angeli
- Unit of Internal Medicine and Hepatology, Department of Medicine, University of Padova, via Giustiniani, 2, 35128, Padova, Italy; European Reference Network - ERN RARELIVER, Department of Medicine, Azienda Ospedale-Università, Padova, Italy
| | - Patrizia Pontisso
- Unit of Internal Medicine and Hepatology, Department of Medicine, University of Padova, via Giustiniani, 2, 35128, Padova, Italy; European Reference Network - ERN RARELIVER, Department of Medicine, Azienda Ospedale-Università, Padova, Italy
| | - Andrea Martini
- Unit of Internal Medicine and Hepatology, Department of Medicine, University of Padova, via Giustiniani, 2, 35128, Padova, Italy; European Reference Network - ERN RARELIVER, Department of Medicine, Azienda Ospedale-Università, Padova, Italy.
| |
Collapse
|
5
|
Yan K, He Q, Tang J, Peng W, Dou B, Chen H, Bei W. Actinobacillus pleuropneumoniae infection activates IL-1β expression in porcine alveolar macrophages via β-amyloid production. Microb Pathog 2025; 204:107559. [PMID: 40220800 DOI: 10.1016/j.micpath.2025.107559] [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: 12/21/2024] [Revised: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Actinobacillus pleuropneumoniae (A. pleuropneumoniae), a porcine respiratory tract pathogen, causes porcine pleuropneumonia. Porcine alveolar macrophages (PAMs) play a crucial role during A. pleuropneumoniae infection. Amyloid precursor protein (APP) can be cleaved by β- and γ-secretase to produce β-amyloid (Aβ). APP and Aβ are associated with the inflammatory response. They activate microglia and astrocytes to secrete IL-1β, IL-6, and other cytokines. In this study, we found that during the interaction between A. pleuropneumoniae and PAMs, the two-component system CpxAR upregulates wecA expression, increasing lipopolysaccharide (LPS) production. LPS promotes APP production and cleavage to generate Aβ. The Aβ activates NF-κB, leading to increased IL-1β expression. We hypothesize that A. pleuropneumoniae infection of PAMs regulates APP production and cleavage to control Aβ levels. Different quantities of Aβ induce PAMs to produce varying amounts of cytokines, leading to different pathological processes in porcine pleuropneumonia.
Collapse
Affiliation(s)
- Kang Yan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Qiyun He
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Jia Tang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Wei Peng
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Beibei Dou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, China
| | - Weicheng Bei
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, China.
| |
Collapse
|
6
|
Wang L, Jiang S, Zhou J, Gholipourmalekabadi M, Cao Y, Lin K, Zhuang Y, Yuan C. From hard tissues to beyond: Progress and challenges of strontium-containing biomaterials in regenerative medicine applications. Bioact Mater 2025; 49:85-120. [PMID: 40124596 PMCID: PMC11928986 DOI: 10.1016/j.bioactmat.2025.02.039] [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: 10/20/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Tissue engineering and regenerative medicine have emerged as crucial disciplines focused on the development of new tissues and organs to overcome the limitations of traditional treatments for tissue damage caused by accidents, diseases, or aging. Strontium ion (Sr2+) has garnered significant attention for its multifaceted role in promoting regeneration medicine and therapy, especially in bone tissue regeneration. Recently, numerous studies further confirm that Sr2+ also plays a critical in soft tissue regeneration. This review firstly summarizes the influence of Sr2+ on critical biological processes such as osteogenesis, angiogenesis, immune modulation, matrix synthesis, mineralization, and antioxidative defence mechanisms. Then details the classification, properties, advantages, and limitations of Sr-containing biomaterials (SrBMs). Additionally, this review extends to the current applications of SrBMs in regenerative medicine for diverse tissues, including bone, cartilage, skeletal muscle, dental pulp, cardiac tissue, skin, hair follicles, etc. Moreover, the review addresses the challenges associated with current SrBMs and provides insights for their future designing and applications in regenerative medicine.
Collapse
Affiliation(s)
- Liyun Wang
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Shengjie Jiang
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Jialiang Zhou
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, 1449614535, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Yuan Cao
- Colorado College, 819 N Tejon Street Box 56, Colorado Springs, 80903, Colorado, USA
| | - Kaili Lin
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Yu Zhuang
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Changyong Yuan
- School of Stomatology, Xuzhou Medical University, Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, 221004, China
| |
Collapse
|
7
|
Wo B, Liu S, Liang Z, Li X. Treatment with human placental extracts inhibits allergic rhinitis by modulating AMPK/SHP1/SHP2/STING signaling. Mol Med Rep 2025; 32:183. [PMID: 40280106 PMCID: PMC12059460 DOI: 10.3892/mmr.2025.13548] [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: 09/03/2024] [Accepted: 03/11/2025] [Indexed: 04/29/2025] Open
Abstract
The present study aimed to investigate the regulatory effects and mechanisms of human placental extracts (HPE) on rats and cell models of ovalbumin (OVA)‑induced allergic rhinitis (AR). IFN‑y and LPS induced AR in vitro. A total of 32 male Sprague‑Dawley (SD) rats were randomly divided into the following four groups: Sham group, model group, model + HPE group and model + HPE + AMPK inhibitor group (n=8 rats/group). With the exception of the sham group, the remaining three groups were sensitized with OVA to establish an AR model, followed by various treatments. Hematoxylin and eosin staining was utilized to observe morphological changes in the nasal mucosa, ELISA was employed to measure serum levels of IL‑1β, interferon (IFN)β, immunoglobulin (Ig)E, IgG1 and IgG2a, and western blotting was conducted to assess protein expression across the groups. The sham group exhibited intact tissue structure with no notable pathological alterations. The model group demonstrated pronounced pathological features, including extensive infiltration of inflammatory cells, tissue shedding and edema. The model + HPE group revealed a gradual restoration of tissue architecture, characterized by reduced edema and inflammatory infiltration, whereas the model + HPE + AMPK inhibitor group again exhibited significant inflammatory cell infiltration and other pathological manifestations. Compared with the sham operation group, the levels of IL‑1β, IFNβ, IgE, IgG1 and IgG2a in the serum of the model group were elevated. The levels of IL‑1β, IFNβ, IgE, IgG1 and IgG2a in the model + HPE group were lower than those in the model group. In addition, the levels of IL‑1β, IFNβ, IgE, IgG1 and IgG2a in the model + HPE + AMPK inhibitor group were higher than those in the model + HPE group. Relative to the sham group, the expression levels of phosphorylated (p)‑AMPK/total (t)‑AMPK, p‑Src homology 2‑containing phosphatase (SHP)1/t‑SHP1 and p‑SHP2/t‑SHP2 were diminished, whereas the expression levels of p‑STING/t‑STING and p‑TBK1/t‑TBK1 were heightened in the model group. In comparison to the model group, the expression levels of p‑AMPK/t‑AMPK, p‑SHP1/t‑SHP1 and p‑SHP2/t‑SHP2 were enhanced, whereas the expression levels of p‑STING/t‑STING and p‑TBK1/t‑TBK1 were reduced in the model + HPE group. Conversely, when compared with the model + HPE group, the expression levels of p‑AMPK/t‑AMPK, p‑SHP1/t‑SHP1 and p‑SHP2/t‑SHP2 were decreased, whereas those of p‑STING/t‑STING and p‑TBK1/t‑TBK1 were increased in the model + HPE + AMPK inhibitor group. In conclusion, HPE may inhibit AR by modulating the AMPK/SHP1/SHP2/STING signaling pathway.
Collapse
Affiliation(s)
- Beibei Wo
- Department of Otolaryngology Head and Neck Surgery, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Shuang Liu
- Department of Pathology, The 980th Hospital of People's Liberation Army (PLA) Joint Logistics Support Force, Shijiazhuang, Hebei 050082, P.R. China
| | - Zihui Liang
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Xiaoming Li
- Department of Otolaryngology Head and Neck Surgery, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
8
|
Liu T, Huang Y, Wang Y, Shen H. Disrupting the immune homeostasis: the emerging role of macrophage ferroptosis in autoimmune diseases. Int Immunopharmacol 2025; 157:114745. [PMID: 40319750 DOI: 10.1016/j.intimp.2025.114745] [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: 12/19/2024] [Revised: 03/18/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Autoimmune diseases are a class of chronic disorders characterized by the aberrant activation of the immune system, where macrophages play a central role in regulating immune responses during disease onset and progression. Ferroptosis, a form of iron-dependent programmed cell death, has recently attracted significant interest due to its involvement in various pathological conditions. In macrophages, ferroptosis not only compromises cell viability but also disrupts immune homeostasis by promoting pro-inflammatory responses and suppressing anti-inflammatory pathways, thereby intensifying inflammation and exacerbating disease severity. While substantial progress has been made in elucidating macrophage ferroptosis in atherosclerosis and oncology, its precise mechanistic role in autoimmune diseases remains largely unexplored. This review systematically summarizes the molecular mechanisms of macrophage ferroptosis and its regulatory effects on immune homeostasis, with particular emphasis on its role in autoimmune diseases, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), inflammatory bowel disease (IBD), multiple sclerosis (MS), and systemic sclerosis (SSc). Furthermore, we discuss potential therapeutic targets related to macrophage ferroptosis in these conditions. By integrating current knowledge, this review aims to provide a theoretical framework and novel perspectives for developing innovative therapeutic strategies targeting autoimmune diseases.
Collapse
Affiliation(s)
- Tianfu Liu
- Department of Hepatology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China
| | - Yichen Huang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China
| | - Yizhe Wang
- Department of Respiratory and Critical Care Medicine, The First People Hospital of Lanzhou, Lanzhou 730050, Gansu, China
| | - Haili Shen
- Department of Rheumatology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, Gansu, China.
| |
Collapse
|
9
|
Liu B, Li Y, Chen H, Li S, Dan X, Xue P, Li Y, Lei L, Fan X. From molecular mechanisms to clinical translation: Silk fibroin-based biomaterials for next-generation wound healing. Int J Biol Macromol 2025; 313:144266. [PMID: 40381758 DOI: 10.1016/j.ijbiomac.2025.144266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Silk fibroin (SF) is a natural polymeric material that has attracted intense research attention in the field of wound healing due to its exceptional mechanical properties, tunable biodegradability, and multifunctional bioactivity. This review systematically summarizes the preparation strategies, functional modifications, and multidimensional application mechanisms of SF and its composite materials in wound healing. The innovative applications of SF in intelligent dressing design, immunometabolic regulation, controlled drug release, stem-cell function modulation, and bioelectrical-activity-mediated microenvironment remodeling is further explored, while analyzing the therapeutic efficacy and cost-effectiveness of SF through clinical translation cases. Distinct from previous reviews, this work not only integrates the latest advances in SF molecular mechanisms and material design but also emphasizes its potential in precision medicine, such as the development of genetically engineered SF for customized immunoregulatory networks. Finally, the article highlights the current challenges in the development of SF materials, including mechanical stability, degradation controllability, and standardization of large-scale production, and envisions future research directions driven by 3D bioprinting and synthetic biology technologies. This review provides a theoretical foundation and technical reference information for the development of efficient, multifunctional, and clinically translatable SF-based materials for application in wound healing.
Collapse
Affiliation(s)
- Bo Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yang Li
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Han Chen
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Songjie Li
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xin Dan
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Ping Xue
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yuli Li
- Shanxi Key Laboratory for Animal Conservation, College of Life Science, Northwest University, Xi'an 710069, China.
| | - Lanjie Lei
- Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China.
| | - Xing Fan
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
10
|
Liu D, Liu L, Zhao X, Zhang X, Chen X, Che X, Wu G. A comprehensive review on targeting diverse immune cells for anticancer therapy: Beyond immune checkpoint inhibitors. Crit Rev Oncol Hematol 2025; 210:104702. [PMID: 40122356 DOI: 10.1016/j.critrevonc.2025.104702] [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: 02/10/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, primary resistance and acquired resistance continue to limit their efficacy for many patients. To address resistance and enhance the anti-tumor activity within the tumor immune microenvironment (TIME), numerous therapeutic strategies targeting both innate and adaptive immune cells have emerged. These include combination therapies with ICIs, chimeric antigen receptor T-cell (CAR-T), chimeric antigen receptor macrophages (CAR-Ms) or chimeric antigen receptor natural killer cell (CAR-NK) therapy, colony stimulating factor 1 receptor (CSF1R) inhibitors, dendritic cell (DC) vaccines, toll-like receptor (TLR) agonists, cytokine therapies, and chemokine inhibition. These approaches underscore the significant potential of the TIME in cancer treatment. This article provides a comprehensive and up-to-date review of the mechanisms of action of various innate and adaptive immune cells within the TIME, as well as the therapeutic strategies targeting each immune cell type, aiming to deepen the understanding of their therapeutic potential.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinming Zhao
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaoman Zhang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaochi Chen
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
11
|
Wu L, He J, Shen N, Chen S. Molecular and cellular mechanisms underlying peripheral nerve injury-induced cellular ecological shifts: Implications for neuroregeneration. IBRO Neurosci Rep 2025; 18:120-129. [PMID: 39877591 PMCID: PMC11773043 DOI: 10.1016/j.ibneur.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
The peripheral nervous system is a complex ecological network, and its injury triggers a series of fine-grained intercellular regulations that play a crucial role in the repair process. The peripheral nervous system is a sophisticated ecological network, and its injury initiates a cascade of intricate intercellular regulatory processes that are instrumental in the repair process. Despite the advent of sophisticated microsurgical techniques, the repair of peripheral nerve injuries frequently proves inadequate, resulting in adverse effects on patients' quality of life. Accordingly, the continued pursuit of more efficacious treatments is of paramount importance. In this paper, a review of the relevant literature from recent years was conducted to identify the key cell types involved after peripheral nerve injury. These included Schwann cells, macrophages, neutrophils, endothelial cells, and fibroblasts. The review was conducted in depth. This paper analyses the phenotypic changes of these cells after injury, the relevant factors affecting these changes, and how they coordinate with neurons and other cell types. In addition, it explores the potential mechanisms that mediate the behaviour of these cells. Understanding the interactions between these cells and their mutual regulation with neurons is of great significance for the discovery of new neuroregenerative treatments and the identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Limao Wu
- School of Clinical Medicine, Hebei University of Engineering, No.81 Congtai Road, Congtai District, Handan City, Hebei Province 056004, China
| | - Jinglan He
- Affiliated Hospital of Hebei University of Engineering, No. 80, Jianshe Street, Fuxing District, Handan City, Hebei Province 056003, China
| | - Na Shen
- Department of Science and Education, Affiliated Hospital of Hebei University of Engineering, No.81 Congtai Road, Congtai District, Handan City, Hebei Province 056004, China
| | - Song Chen
- Orthopaedic Center, Affiliated Hospital of Hebei University of Engineering, No.81 Congtai Road, Congtai District, Handan City, Hebei Province 56004, China
| |
Collapse
|
12
|
Fang B, Lu Y, Li X, Wei Y, Ye D, Wei G, Zhu Y. Targeting the tumor microenvironment, a new therapeutic approach for prostate cancer. Prostate Cancer Prostatic Dis 2025; 28:260-269. [PMID: 38565910 DOI: 10.1038/s41391-024-00825-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND A growing number of studies have shown that in addition to adaptive immune cells such as CD8 + T cells and CD4 + T cells, various other cellular components within prostate cancer (PCa) tumor microenvironment (TME), mainly tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs) and myeloid-derived suppressor cells (MDSCs), have been increasingly recognized as important modulators of tumor progression and promising therapeutic targets. OBJECTIVE In this review, we aim to delineate the mechanisms by which TAMs, CAFs and MDSCs interact with PCa cells in the TME, summarize the therapeutic advancements targeting these cells and discuss potential new therapeutic avenues. METHODS We searched PubMed for relevant studies published through December 10 2023 on TAMs, CAFs and MDSCs in PCa. RESULTS TAMs, CAFs and MDSCs play a critical role in the tumorigenesis, progression, and metastasis of PCa. Moreover, they substantially mediate therapeutic resistance against conventional treatments including anti-androgen therapy, chemotherapy, and immunotherapy. Therapeutic interventions targeting these cellular components have demonstrated promising effects in preclinical models and several clinical trials for PCa, when administrated alone, or combined with other anti-cancer therapies. However, the lack of reliable biomarkers for patient selection and incomplete understanding of the mechanisms underlying the interactions between these cellular components and PCa cells hinder their clinical translation and utility. CONCLUSION New therapeutic strategies targeting TAMs, CAFs, and MDSCs in PCa hold promising prospects. Future research endeavors should focus on a more comprehensive exploration of the specific mechanisms by which these cells contribute to PCa, aiming to identify additional drug targets and conduct more clinical trials to validate the safety and efficacy of these treatment strategies.
Collapse
Affiliation(s)
- Bangwei Fang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Ying Lu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaomeng Li
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Gonghong Wei
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| |
Collapse
|
13
|
Li H, Chen J, Guo H, Yang H, Liu J, Yuan H, Zhang J, Wang J, Liu S. Integrated transcriptomic and proteomic profiling reveals the anti-inflammatory mechanism of dihydroartemisinin in the treatment of acute liver injury by targeting CYBA and CYBB. Biochem Biophys Res Commun 2025; 764:151821. [PMID: 40250321 DOI: 10.1016/j.bbrc.2025.151821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Acute liver injury (ALI) is a prevalent inflammatory disease with no currently available effective targeted therapies that characterized by high mortality and morbidity. Dihydroartemisinin (DHA), a derivative of the renowned antimalarial compound artemisinin, has garnered attention for its anti-inflammatory property. However, the precise anti-inflammatory mechanisms underlying its efficacy in treating ALI remain unclear. Notably, the excessive inflammatory cytokines secreted by macrophages represents a critical factor of liver damage. In our comprehensive study, transcriptome and proteomic analysis of M1 macrophages after DHA treatment was performed to unearth the potential anti-inflammatory targets for ALI treatment. Transcriptomics analysis indicated that DHA significantly mitigated inflammation, primarily by downregulating the expressions of CCL1, CCL2, CCL7, CCL13, and CXCL13. Concurrently, proteomics analysis identified six proteins, such as CYBA and CYBB, that were consistently downregulated in the DHA intervention groups compared to the M1 group. Intriguingly, a protein-protein interaction network analysis highlighted the close association of CYBA and CYBB with the aforementioned chemokine genes. Through meticulous screening, DHA curtailed the production of reactive oxygen species (ROS) by targeting CYBA and CYBB, subsequently suppressing the secretion of several chemokines and dampening the inflammatory response in M1 macrophages. More importantly, DHA not only reduced ROS and chemokine levels but also restored liver function by downregulating CYBA and CYBB to inhibit NF-κB pathway in ALI mice, demonstrating strong anti-inflammatory effects. In conclusion, our findings throw novel light into the underlying anti-inflammatory mechanism of DHA in ALI management, offering valuable insights for future clinical research and therapeutic strategies for inflammatory diseases.
Collapse
Affiliation(s)
- Honglian Li
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiayun Chen
- State Key Laboratory for Quality Ensuanse and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Huiyi Guo
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hao Yang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jing Liu
- State Key Laboratory for Quality Ensuanse and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Haoxing Yuan
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensuanse and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jigang Wang
- State Key Laboratory for Quality Ensuanse and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou, 510515, China; MOE Innovation Center for Medical Basic Research on Inflammation and Immune Related Diseases, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
14
|
Zhu Q, Chen Z, Wang D, Jiao X, Luan Y, Wang M, Luo R, Wang Y, Fu G, Wang Y, Zhang W. Microenvironment-responsive coating for vascular stents to regulate coagulation-inflammation interaction and promote vascular recovery. Bioact Mater 2025; 48:443-457. [PMID: 40093305 PMCID: PMC11909720 DOI: 10.1016/j.bioactmat.2025.02.031] [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: 10/27/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
Early coagulation-inflammation interaction and late in-stent restenosis undermine the efficacy of vascular stents after implantation. Targeting the interplay between inflammation and coagulation, and smooth muscle cell (SMC) proliferation, we presented a microenvironment-responsive coating designed to regulate tissue responses and vascular regeneration throughout the remodeling process. Coagulation was inhibited by incorporating anticoagulant tirofiban into the coating. MMP9-responsive nanoparticles embedded in the coating released salvianolic acid A to modulate inflammatory cell behavior and inhibit SMC dysfunction. By effectively interfering with clotting and inflammation, the coating suppressed platelet-fibrin interaction and formation of platelet-monocyte aggregates, thereby mitigating adverse effects on reendothelialization. Its ability to influence SMC proliferation and migration resulted in reduced intimal hyperplasia. Coated stents were shown to significantly regulate tissue regeneration, improve the vascular environment and even reduced the lipid content in the narrowed atherosclerotic vessels in vivo. This direct approach enhanced the vascular tissue regeneration after stent implantation, and offered promising insights for optimizing vascular stent design.
Collapse
Affiliation(s)
- Qiongjun Zhu
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Zhezhe Chen
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Dan'an Wang
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Xiaolu Jiao
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Yi Luan
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Min Wang
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Yanan Wang
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Wenbin Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| |
Collapse
|
15
|
Liang W, Wu H, Long Q, Lin H, Lv X, Ma W, Wu T, Li A, Zheng Q, Guo J, Chen X, Guo J, Sun D. LKB1 activated by NaB inhibits the IL-4/STAT6 axis and ameliorates renal fibrosis through the suppression of M2 macrophage polarization. Life Sci 2025; 370:123564. [PMID: 40097066 DOI: 10.1016/j.lfs.2025.123564] [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: 12/07/2024] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Renal fibrosis is a critical pathological characteristic of chronic kidney disease, and current antifibrotic therapies has limited efficacy. Sodium butyrate (NaB) has been shown to be highly effective in mitigating bleomycin-induced pulmonary fibrosis; however, its specific impact on renal fibrosis and the underlying mechanisms remain unclear. This study aims to elucidate the role and mechanism of NaB in renal fibrosis by using a mouse model of renal fibrosis induced through Unilateral Ureteral Obstruction (UUO) and folic acid (FA) administration. RESULTS NaB significantly decreased the distribution of collagen fibers in renal tissues and mitigated fibrosis in a dose-dependent manner. Further analysis indicated that NaB inhibited M2 macrophage polarization in the renal tissues of UUO model mice by blocking the phosphorylation of STAT6, hence reducing renal fibrosis. Additionally, in vitro experiments demonstrated that NaB inhibited fibroblast activation induced by M2 macrophages. Mechanistic studies revealed that NaB attenuates fibroblast activation and M2 macrophage polarization by upregulating LKB1 and inhibiting the activation of the STAT6 signaling pathway. CONCLUSION NaB may exert its effects by inhibiting the activation of the IL-4/STAT6 signaling pathway through the upregulation of LKB1, which suppress the polarization of M2 macrophages and consequently reduce renal fibrosis. These findings establish a theoretical foundation for NaB as a novel drug candidate for renal fibrosis and indicate its potential applicability in clinical treatments for this condition.
Collapse
Affiliation(s)
- Weifei Liang
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China; Center for Cancer and Immunology Research, State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, 510180 Guangzhou, Guangdong, China
| | - Haoyu Wu
- School of Public Health, Wenzhou Medical University, Wenzhou 325035, China; South Zhejiang Institute of Radiation Medicine and Nuclear Technology Application, Wenzhou 325809, China; Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Qishan Long
- Department of Urology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Hong Lin
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, 511518 Qingyuan, Guangdong, China
| | - Xiaoyu Lv
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Xuhui District, Shanghai 200032, China
| | - Wen Ma
- Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China
| | - Tao Wu
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China
| | - Ai Li
- Department of Clinical Medicine, The Second Clinical School of Guangzhou Medical University, Guangzhou 510000, China
| | - Qingyou Zheng
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China
| | - Jinan Guo
- Department of Urology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China.
| | - Xiangqiu Chen
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China.
| | - Jing Guo
- Center of Oncology, Heyou Hospital, Shunde District, Foshan City 528306, Address:No. 1 of Heren Road, Junlan Community, Beijiao Town, Shunde District, Foshan City, Guangdong Province, China.
| | - Donglin Sun
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China.
| |
Collapse
|
16
|
Jia L, Wang Y, Jin C, Ma Y, Wang Y, Song L, Shen J, Xie Y, Xiang M. Macrophage heme oxygenase-1 modulates peroxynitrite-mediated vascular injury and exacerbates abdominal aortic aneurysm development. Am J Physiol Cell Physiol 2025; 328:C1808-C1821. [PMID: 40261277 DOI: 10.1152/ajpcell.00525.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/01/2023] [Accepted: 04/11/2025] [Indexed: 04/24/2025]
Abstract
Inflammatory reactions mediated by macrophages are profoundly related to the depletion of smooth muscle cells (SMCs) in abdominal aortic aneurysm (AAA) development. The findings from our previous investigation indicate that heme oxygenase-1 (HO-1) in macrophages exacerbates proinflammatory responses and oxidative damage. Therefore, the aim of this work was to gain insight into the function of HO-1 derived from macrophages and elucidate the underlying molecular mechanisms involved in AAA development. In this study, we discovered a dramatic increase in HO-1 expression in the infiltrated macrophages in experimental calcium phosphate-induced AAA tissues. Myeloid conditional HO-1-deficient mice displayed slower luminal area enlargement, as well as diminished inducible nitric oxide synthase (iNOS)-positive M1 macrophage activation, peroxynitrite generation, and SMCs apoptosis in aneurysmal tissues compared with littermate controls. Furthermore, we showed that inhibiting HO-1 eliminated the protein expression of iNOS induced by lipopolysaccharide/interferon-γ in bone marrow-derived macrophages, whereas the mRNA expression remained unaffected. Suppressing iNOS in macrophages alleviated SMCs apoptosis by decreasing nitric oxide generation in a coculture system in vitro. In summary, our study illustrates that macrophage-derived HO-1 strengthens AAA development through boosting the production of iNOS-dependent peroxynitrite and the deterioration of SMCs. These findings reveal potential therapeutic targets for resolving aneurysmal diseases.NEW & NOTEWORTHY This article illustrates the role of macrophage-derived heme oxygenase-1 (HO-1) in the development of abdominal aortic aneurysm (AAA). HO-1 deletion in macrophages hindered AAA development by reducing inducible nitric oxide synthase (iNOS)-dependent peroxynitrite production and smooth muscle cells (SMCs) apoptosis in vivo. Mechanistically, inhibition of HO-1 reduced the stimulated iNOS protein production in macrophages by lipopolysaccharide/interferon-γ. Moreover, suppressing iNOS in macrophages prevented SMCs apoptosis by decreasing nitric oxide generation in vitro.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/enzymology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Macrophages/enzymology
- Macrophages/pathology
- Peroxynitrous Acid/metabolism
- Nitric Oxide Synthase Type II/metabolism
- Nitric Oxide Synthase Type II/genetics
- Apoptosis
- Mice
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- Heme Oxygenase-1/metabolism
- Heme Oxygenase-1/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Male
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Disease Models, Animal
- Aorta, Abdominal/pathology
- Aorta, Abdominal/enzymology
- Nitric Oxide/metabolism
- Membrane Proteins
Collapse
Affiliation(s)
- Liangliang Jia
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Yufei Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Chunna Jin
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Yuankun Ma
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Yidong Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Liuguang Song
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Jian Shen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, People's Republic of China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
17
|
Wang X, Song L, Zhao J, Xiong Y, Jin R, He J. Matrix viscoelasticity drives cell cluster formation to counteract cellular senescence. J Mater Chem B 2025. [PMID: 40432619 DOI: 10.1039/d5tb00174a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
During tissue repair, stress-induced cellular senescence represents a critical factor that impedes the regenerative potential of tissues. While the regulatory effects of matrix viscoelasticity on cellular behavior have been documented, their role and correlated mechanisms underlying cellular senescence remain unclear. In this study, we engineered a viscoelastic gel matrix exhibiting a storage modulus of approximately 3 kPa, with a tunable loss modulus ranging from 0 to 300 Pa by incorporating linear alginate and modulating the compactness of a polyacrylamide-based covalent network. Utilizing a UV-induced senescence model, we observed that increasing the matrix's viscoelasticity from 0 Pa to 300 Pa led to a significant reduction in the proportion of senescent cells, from 90.5% to 22.7%. Furthermore, cells cultured in these matrices exhibited a tendency to form cell aggregation, with the cell populations demonstrating a collective resistance to stresses. This indicated that viscoelastic materials would promote enhanced cellular interactions, thereby strengthening cellular resilience against UV-induced stresses. Furthermore, combined with microarray analysis, it was concluded that the presence of viscoelastic components activated the connexin 43 (Cx43)-modulated gap junction for cluster formation, thereby suppressing the senescence-associated signaling pathways, including Wnt/β-catenin, MAPK, NF-κB, and TGF-β. Additionally, the integrin-cytoskeleton-Yes-associated protein (YAP) signaling axis played an active role in delaying cell aging. These results provide novel insights into the regulatory role of viscoelastic materials in cellular senescence and offer a compelling foundation for the development of advanced biomaterials for tissue repair.
Collapse
Affiliation(s)
- Xinying Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China.
| | - Limin Song
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China.
| | - Jingwen Zhao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China.
| | - Yiling Xiong
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China.
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China.
| | - Jing He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China.
| |
Collapse
|
18
|
Li Z, Liu L, Sun Y, Liu X, Zhang P, Wang Y, Ding G. Mesenchymal stem/stromal cells-derived exosomes: possible therapeutic mechanism in inflammatory bowel disease. Hum Cell 2025; 38:111. [PMID: 40434563 DOI: 10.1007/s13577-025-01243-6] [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: 04/02/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract caused by dysfunction of the immune system in genetically susceptible individuals. As current pharmacologic and surgical treatments remain suboptimal, increasing attention has been directed toward exosomes derived from mesenchymal stem/stromal cells (MSCs) as alternative therapeutic approaches. MSCs are multipotent stromal cells that can be isolated from various human tissues such as bone marrow, adipose, umbilical cord and periodontal ligament. Exosomes are cell-derived membrane-bound vesicles enclosing RNAs, proteins, growth factors, and cytokines. Previous studies indicate that the anti-inflammatory, immunomodulatory, and regenerative effects of MSCs are largely mediated by MSC-derived exosomes (MSC-Exos). Therefore, this review outlines current insights into the molecular mechanisms of MSC-Exos in IBD treatment to support the future development of MSC-Exos as a therapeutic strategy, thus providing novel observations into the clinical applications of MSC-Exos in IBD management.
Collapse
Affiliation(s)
- Zekun Li
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Luyun Liu
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Yuhui Sun
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Xinjuan Liu
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Ping Zhang
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Yue Wang
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Gang Ding
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China.
| |
Collapse
|
19
|
Liu X, Jiang R, Xu Y, Xu X, Fang L, Gao G, Han L, Chen Y, Du H, Cai Y, Zhu F, Chen M, Wang K, Li H, Wang G, Quan C. Dual cytokine-engineered macrophages rejuvenate the tumor microenvironment and enhance anti-PD-1 therapy in renal cell carcinoma. Int Immunopharmacol 2025; 156:114725. [PMID: 40294469 DOI: 10.1016/j.intimp.2025.114725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025]
Abstract
Despite advances in PD-1 blockade therapy, the immunosuppressive tumor microenvironment (TME) limits its efficacy in renal cell carcinoma (RCC). Here, we developed dual-cytokine-engineered macrophages co-delivering IL-12 and CXCL-9 to reprogram TME and enhance anti-PD-1 responsiveness. Single-cell RNA sequencing revealed that RCC harbor abundant M2-like tumor-associated macrophages (TAMs), which correlate with T-cell exhaustion. In vitro, engineered macrophages polarized M2-like TAMs to antitumor M1 phenotypes, secreted CXCL-9 to recruit cytotoxic T cells, and released IL-12 to amplify T/NK cell activation. In vivo, intravenously administered engineered macrophages homed to tumors, reshaped the TME by increasing CD8+ T cells, dendritic cells, and NK cells while reducing immunosuppressive Tregs and MDSCs. This approach synergized with PD-1 blockade, resulting in a 2.5-fold greater tumor growth inhibition compared to anti-PD-1 monotherapy. This dual-cytokine macrophage platform offers a novel strategy to overcome resistance to checkpoint inhibitors in RCC by delivering cytokine and remodeling TME, with implications for clinical translation.
Collapse
Affiliation(s)
- Xin Liu
- Tianjin institute of urology,Tianjin Medical University Second Hospital, Tianjin, China; Department of Urology,The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ranran Jiang
- Department of Oncology,The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yujun Xu
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaodi Xu
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lin Fang
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ge Gao
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lulu Han
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuxin Chen
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongwei Du
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ying Cai
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fei Zhu
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingjing Chen
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kaidi Wang
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hailong Li
- Department of Urology,The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Gang Wang
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Changyi Quan
- Tianjin institute of urology,Tianjin Medical University Second Hospital, Tianjin, China.
| |
Collapse
|
20
|
Zhu S, Liu J, Xu K, Xu F, Jiang Y, Dai L, Pei T, Zhu Y, Liu D, Zhang X, Xu J, Yang J, Pan Z, Tao J, Hou Z. Comparative transcriptomic analyses of macrophages infected with Toxoplasma gondii strains of different virulence provide molecular insights into the response of macrophage in phagocytosis and polarization to infection. Mol Immunol 2025; 183:259-273. [PMID: 40414092 DOI: 10.1016/j.molimm.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/07/2025] [Accepted: 05/05/2025] [Indexed: 05/27/2025]
Abstract
Macrophages are essential for the proliferation and spread of Toxoplasma gondii. Modulating macrophage activation to improve the inflammatory environment is an effective approach for disease treatment. However, the molecular mechanism through which T. gondii alters macrophage function remain unknown. Based on transcriptomic data analysis of various macrophage types infected with T. gondii, current research revealed differences in the regulation of macrophage functions among strains with different virulence: RH was primarily involved in cell cycle regulation, ME49 was associated with cAMP signaling, and CEP mainly participated in ion channel activity. All three T. gondii strains were involved in regulating immune response activation, including leukocyte adhesion and the MAPK signaling pathway. Nineteen shared DEGs associated with macrophage phagocytosis or polarization were identified through the GeneCards database, and PPI analysis confirmed Il6 as the hub gene in the regulatory network. In vivo and in vitro experiments showed that the YZ-1 strain significantly regulated the expressions of eight DEGs (Il6, Rel, Cd83, Myc, Adora2b, Egr2, Gja1 and Nr4a2), and promoted macrophage phagocytic activity and induced M1 polarization, confirming a significant correlation with Il6. This study revealed the dissimilarities and commonalities in macrophage function regulated by T. gondii strains of different virulence, and identified key molecules involved in the regulation of macrophage phagocytosis and polarization during T. gondii infection. This is crucial for identifying potential drug targets against T. gondii and provides a new perspective on the etiopathogenesis and therapeutic approaches for toxoplasmosis.
Collapse
Affiliation(s)
- Shifan Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Jiantao Liu
- YEBIO Bioengineering Co., Ltd of QINGDAO, Qingdao 266113, PR China
| | - Kangzhi Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Fan Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Yuwei Jiang
- Lingkou Town Animal Epidemic Prevention Station, Danyang 212353, PR China
| | - Linwei Dai
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Tianxu Pei
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Yuyang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Dandan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Xinjun Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Jinjun Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Jin Yang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225000, PR China.
| | - Zhiming Pan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China
| | - Jianping Tao
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China.
| | - Zhaofeng Hou
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou 225009, PR China; International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, PR China.
| |
Collapse
|
21
|
Martkamjan C, Lerdsudkanung K, Tipay PS, Rezgui R, Teo JCM, Sapudom J. Machine learning-based label-free macrophage phenotyping in immune-material interactions. J Mater Chem B 2025; 13:5858-5870. [PMID: 40289902 DOI: 10.1039/d5tb00365b] [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: 04/30/2025]
Abstract
The rapid advancement of implantable biomedical materials necessitates a comprehensive understanding of macrophage interactions to optimize implant immunocompatibility. Macrophages, key immune regulators, exhibit phenotypic plasticity by polarizing into pro-inflammatory (M1) or anti-inflammatory (M2) subtypes. Conventional phenotyping techniques, such as flow cytometry and immunostaining, provide insights but have limitations related to fixation and endpoint analysis. This study presents a high-throughput, label-free macrophage phenotyping approach integrating AI-driven image classification with quantitative phase imaging (QPI). THP-1-derived macrophages were differentiated into M0, M1, M2a, and M2c phenotypes, and their morphological and refractive index properties were analyzed using QPI. Although QPI alone could not fully distinguish phenotypes, deep learning models, including GoogLeNet, ShuffleNet, VGG-16, and ResNet-18, were evaluated, with ResNet-18 achieving over 90% accuracy. Additionally, macrophage responses to collagen coatings (types I, III, and IV) were assessed using machine learning-based phenotyping and cytokine profiling. Collagen I induced an M1 response, collagen III supported a balanced M1/M2 profile, and collagen IV promoted a controlled immune environment. These findings demonstrate the potential of AI-driven QPI as a non-invasive tool for macrophage characterization, offering insights into biomaterial immunocompatibility and informing implant design strategies.
Collapse
Affiliation(s)
- Chawalwat Martkamjan
- International School of Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok, Thailand
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
| | - Kornlavit Lerdsudkanung
- International School of Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok, Thailand
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
| | - Paul Sean Tipay
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
- Independent Researcher, Abu Dhabi, United Arab Emirates
| | - Rachid Rezgui
- Core Technology Platform - Light Microscopy, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Jeremy C M Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
22
|
Chen S, Ciccone N, Ray D. The clockwork macrophage: timing in innate immunity. Biomed J 2025:100872. [PMID: 40409505 DOI: 10.1016/j.bj.2025.100872] [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: 01/02/2025] [Revised: 04/30/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025] Open
Abstract
The circadian clock enables organisms to predict daily environmental changes and synchronize their physiology and behaviour accordingly. Macrophages, key sensor cells in the innate immune system, exhibit cell-autonomous circadian rhythmicity. This circadian rhythmic behaviour is synchronised to the central clock in the hypothalamus as a result of neural, and hormonal signals. Macrophage rhythms and responses involve sensing temporal cues, integrating information from tissue-specific environments, and initiating context-appropriate, time-gated responses. On a broader scale, monocytes and macrophages communicate and synchronize with other immune cells, migrate throughout the body, and infiltrate tissues, collectively contributing to circadian regulation in both health and disease. While the field of macrophage circadian biology is rapidly advancing, it is equally important to reflect on its historical development, which has been shaped by over two centuries of accumulating knowledge and technological progress. This review traces key milestones in macrophage and circadian research, examining how recent discoveries have refined our understanding of early foundational questions and setting the stage for future inquiries. Notably, many intriguing questions remain unresolved, including the circadian regulation of macrophage function under steady-state conditions, the tissue-specific heterogeneity of macrophage circadian rhythms, and the role of macrophage circadian clocks in disease pathogenesis and their potential clinical implications.
Collapse
Affiliation(s)
- Siyu Chen
- NIHR Oxford Health Biomedical Research Centre, and NIHR; Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, and Oxford Kavli Centre for Nanoscience Discovery, University of Oxford, Oxford, OX3 7LE, UK
| | - Nick Ciccone
- NIHR Oxford Health Biomedical Research Centre, and NIHR; Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, and Oxford Kavli Centre for Nanoscience Discovery, University of Oxford, Oxford, OX3 7LE, UK
| | - David Ray
- NIHR Oxford Health Biomedical Research Centre, and NIHR; Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK; Oxford Centre for Diabetes, Endocrinology and Metabolism, and Oxford Kavli Centre for Nanoscience Discovery, University of Oxford, Oxford, OX3 7LE, UK.
| |
Collapse
|
23
|
Li R, Shu Y, Yan Y, Zhu J, Cheng Z, Zhang J, Zhu L, Qiao Y, Sun Q. Caveolin-1 Deficiency in Macrophages Alleviates Carbon Tetra-Chloride-Induced Acute Liver Injury in Mice. Int J Mol Sci 2025; 26:4903. [PMID: 40430042 PMCID: PMC12112502 DOI: 10.3390/ijms26104903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/08/2025] [Accepted: 05/11/2025] [Indexed: 05/29/2025] Open
Abstract
Bone marrow-derived macrophages (BMMs) exhibit dynamic behavior and functional capabilities in response to specific microenvironmental stimuli. Recent investigations have proved that BMMs play crucial roles in promoting necrotic lesion resolution. Despite substantial advancements in understanding their activation and interaction with injured livers, researchers face challenges to develop effective treatments based on manipulating BMMs function. Caveolin-1 (Cav-1) is the major structural protein on the plasma membrane. We previously reported that Cav-1 knockout (KO) mice exhibited less functional damage and necrosis in carbon tetrachloride (CCl4)-induced liver injury. We hypothesize that the activation and recruitment of BMMs are involved in the resolution of necrotic lesions in Cav-1 KO mice. Wild-type (WT) and Cav-1 KO mice were injected with CCl4 (10% v/v) to induce acute liver injury model. Blood samples and hepatic tissues were harvested for serum alanine transaminase (ALT) activity assessment, histopathological examination through hematoxylin-eosin (H&E) staining, and BMMs subpopulation analysis via flow cytometry. Then, primary BMMs were isolated and cultured to investigate the effect of Cav-1 on BMMs polarization, migration, and activation of STAT3 signal pathway. Validation of hepatic macrophage depletion was induced by administrating clodronate liposomes (CLs), and BMMs reconstitution was evaluated by EGFP labelled BMMs. Following this, hepatic macrophages were depleted by CLs, BMMs were isolated from Cav-1 KO, and WT mice were cultured and administrated to evaluate the protective role of Cav-1-deleted BMMs on the resolution of hepatocellular necrosis and apoptosis in acute liver injury. The BMMs ratio significantly increased from 2.12% (1D), 4.38% (1W), and 5.38% (2W) in oil control mice to 7.17%, 14.90%, and 19.30% in CCl4-treated mice (p < 0.01 or p < 0.001). Concurrently, Cav-1 positive BMMs exhibited a marked elevation from 6.41% at 1D to 24.90% by 2W (p = 0.0228). Cav-1 KO exerted protective effects by reducing serum ALT by 26% (p = 0.0265) and necrotic areas by 28% (p = 0.0220) and enhancing BMMs infiltration by 60% (p = 0.0059). In vitro, Cav-1 KO BMMs showed a decrease in CD206 fluorescence intensity (p < 0.001), a time-dependent upregulation of arginase-1 mRNA (p < 0.05 or p < 0.01), a 1.22-fold increase in phosphorylated STAT3 (p = 0.0036), and impaired wound healing from 12 to 24 h (p < 0.001). The macrophage-depleting action in livers by CL injection persists for a minimum of 48 h. Administrated EGFP+ BMMs emerged as the predominant population following CL injection for a duration of 48 h. Following clodronate liposome-mediated hepatic macrophage depletion, the adoptive transfer of Cav-1 KO BMMs demonstrated therapeutic efficacy in CCl4-induced acute liver injury. In CCl4-induced acute liver injury, the adoptive transfer of Cav-1 KO BMMs reduced necrosis by 12.8% (p = 0.0105), apoptosis by 25.2% (p = 0.0127), doubled macrophages infiltration (p = 0.0269), and suppressed CXCL9/10 mRNA expression (p = 0.0044 or p = 0.0385). BMMs play a key role in the resolution of liver necrotic lesions in CCl4-induced acute liver injury. Cav-1 depletion attenuates hepatocellular necrosis and apoptosis by accelerating BMMs recruitment and M2 polarization. Cav-1 in macrophages may represent a potential therapeutic target for acute liver injury.
Collapse
Affiliation(s)
- Ruirui Li
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (R.L.); (Y.S.); (Y.Y.); (J.Z.); (Z.C.)
| | - Yixue Shu
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (R.L.); (Y.S.); (Y.Y.); (J.Z.); (Z.C.)
| | - Yulin Yan
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (R.L.); (Y.S.); (Y.Y.); (J.Z.); (Z.C.)
| | - Junyi Zhu
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (R.L.); (Y.S.); (Y.Y.); (J.Z.); (Z.C.)
| | - Zilu Cheng
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (R.L.); (Y.S.); (Y.Y.); (J.Z.); (Z.C.)
| | - Jie Zhang
- Laboratory Animal Center, Capital Medical University, Beijing 100069, China; (J.Z.); (L.Z.); (Y.Q.)
| | - Liming Zhu
- Laboratory Animal Center, Capital Medical University, Beijing 100069, China; (J.Z.); (L.Z.); (Y.Q.)
| | - Yanhua Qiao
- Laboratory Animal Center, Capital Medical University, Beijing 100069, China; (J.Z.); (L.Z.); (Y.Q.)
| | - Quan Sun
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (R.L.); (Y.S.); (Y.Y.); (J.Z.); (Z.C.)
- Laboratory Animal Center, Capital Medical University, Beijing 100069, China; (J.Z.); (L.Z.); (Y.Q.)
| |
Collapse
|
24
|
Yingming W, Jing G, Tianhong W, Zhenyu W. M2 Macrophages Mitigate Ocular Surface Inflammation and Promote Recovery in a Mouse Model of Dry Eye. Exp Eye Res 2025:110439. [PMID: 40403951 DOI: 10.1016/j.exer.2025.110439] [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: 09/03/2024] [Revised: 04/09/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
Dry eye disease (DED) is a chronic, progressive, multifactorial condition characterized by tear film instability and ocular surface damage. Ocular surface inflammation, triggered by multiple pathogenic factors, represents one of the key mechanisms in DED pathogenesis. This study aims to investigate the therapeutic effects of anti-inflammatory M2 macrophages conditioned medium (M2-CM) on ocular surface inflammation and their potential mechanisms in improving dry eye symptoms in a mouse model. Mouse macrophages (RAW264.7) were polarized into M2 macrophages by IL-4 under different osmolarities, and M2-CM was collected. Flow cytometry and ELISA were applied to measure the cytokine expression of the M2 macrophages. Primary mouse corneal epithelial cells (CECs) were co-cultured with RAW264.7 and M2 macrophages using a Transwell system. The viability and migration of CECs were assessed using CCK-8 and scratch assays. Mouse DED was established by subcutaneous injection of scopolamine, and the therapeutic effects of M2-CM were evaluated by phenol red thread test, fluorescein staining, and tear film breakup time (TBUT). PCR and immunofluorescence staining were applied to observe inflammatory factors and cells on the ocular surface. M2 macrophages enhanced CEC viability, proliferation, and migration, but hyperosmolarity inhibited M2 macrophage polarization. In the DED model, M2-CM improved ocular surface conditions, reduced pro-inflammatory cytokine expression, and increased anti-inflammatory factors. Immunofluorescence revealed reduced pro-inflammatory cells (M1 macrophages, Th1, and Th17) and increased M2 macrophages in the ocular tissues after M2-CM treatment. These results suggest that M2-CM ameliorates ocular surface inflammation and promotes recovery in DED, offering a potential therapeutic strategy for DED.
Collapse
Affiliation(s)
- Wang Yingming
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University
| | - Gao Jing
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University
| | - Wu Tianhong
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University
| | - Wang Zhenyu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University.
| |
Collapse
|
25
|
McGill CJ, White OS, Lu RJ, Sampathkumar NK, Benayoun BA. Sex-dimorphic gene regulation in murine macrophages across niches. Immunol Cell Biol 2025. [PMID: 40390161 DOI: 10.1111/imcb.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/21/2025]
Abstract
Macrophages are a key cell type of the innate immune system and are involved at all steps of inflammation: (i) they present antigens to initiate inflammation, (ii) they clear up foreign bodies through phagocytosis and (iii) they resolve inflammation by removing or deactivating mediator cells. Many subtypes of macrophages have been identified, classified by their niche and/or embryonic origin. In order to better develop therapies for conditions with macrophage dysfunction, it is crucial to decipher potential sex differences in key physiological mediators of inflammation so that treatment efficacy can be ensured regardless of biological sex. Here, we conduct a meta-analysis approach of transcriptomics data sets for male vs. female mouse macrophages across 8 niches to characterize conserved sex-dimorphic pathways in macrophages across origins and niches. For this purpose, we leveraged new and publicly available RNA-sequencing data sets from murine macrophages, preprocessed these datasets and filtered them based on objective QC criteria, and performed differential gene expression analysis using sex as the covariate of interest. Differentially expressed (DE) genes were compared across data sets and macrophage subsets, and functional enrichment analysis was performed to identify sex-specific functional differences. Consistent with their presence on the sex chromosomes, three genes were found differentially expressed across datasets (i.e. Xist, Eif2s3y and Ddx3y). More broadly, we found that female-biased pathways across niches are more consistent than male-biased pathways, specifically relating to the extracellular matrix. Our findings increase our understanding of transcriptional similarities across macrophage niches and underscore the importance of including sex as a biological variable in immune-related studies.
Collapse
Affiliation(s)
- Cassandra J McGill
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Olivia S White
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Quantitative and Computational Biology Department, USC Dornsife College of Letters, Arts, and Sciences, Los Angeles, CA, USA
| | - Ryan J Lu
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Nirmal K Sampathkumar
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, CA, USA
- Biochemistry and Molecular Medicine Department, USC Keck School of Medicine, Los Angeles, CA, USA
- USC Norris Comprehensive Cancer Center, Epigenetics and Gene Regulation, Los Angeles, CA, USA
- USC Stem Cell Initiative, Los Angeles, CA, USA
| |
Collapse
|
26
|
Meer A, Mathews A, Cabral M, Tarabokija A, Carroll E, Chaudhry H, Paszek M, Radecker N, Palaia T, de Guzman HC, de Guzman RC. Biocompatibility and wound-healing prospect of KAPs-depleted residual hair biomaterial. Biomater Sci 2025. [PMID: 40387482 DOI: 10.1039/d4bm00777h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
This work is an in-depth investigation of the in vitro and in vivo biocompatibility of processed and treated residual human hair samples with intact cuticle layers. The specimens included oxidized hair with minimal melanin (BLH) and hair with medium- (M-KAP) and low- (L-KAP) amounts of keratin associated proteins (KAPs), confirmed through gel electrophoresis, electron microscopy, trichrome histological staining, and tensile biomechanics, in comparison to the untreated regular hair (REG) control. All hair groups, high KAPs (H-KAPs: REG and BLH), M-KAP, and L-KAP, are non-cytotoxic in the adipose fibroblast's response to their extracts based on the ISO 10993-5 medical device biomaterial testing standard. In vivo mouse subcutaneous implantation (ISO 10993-6, local effects) at 2 weeks showed a foreign body response (FBR) with thin fibrous encapsulation at 28% relative skin dermis thickness; but the L-KAP implant mitigated a significant decrease in FBR area compared to H-KAPs and a lower number of immune cells of mostly macrophages and mast cells on the biomaterial's surface. In the bulk of the capsules, blood vessels and collagen extracellular matrix densities were similar among groups. These findings suggest that small globular KAPs diffuse out of the cortex to the host-biomaterial interface which induce a slightly-elevated FBR but limited to the implant's surface vicinity. For translatability, we evaluated the effectiveness of the residual hair with the most depleted KAPs (L-KAP) in a 10 mm-diameter, splinted, and full-thickness mouse skin excision wound. Treatment with the L-KAP mesh exhibited an 8% healing improvement per day compared to the untreated control: significantly reducing the projected complete healing time by 30%. On-going research focuses on purer keratin-based and macromolecularly organized residual hair biomaterials for drug-delivery as they are deemed the most biocompatible.
Collapse
Affiliation(s)
- Allison Meer
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY 11549, USA..
- Department of Biology, Hofstra University, Hempstead, NY 11549, USA
| | - Aidan Mathews
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY 11549, USA..
- Department of Biology, Hofstra University, Hempstead, NY 11549, USA
| | - Mariana Cabral
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY 11549, USA..
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Andrew Tarabokija
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY 11549, USA..
| | - Evan Carroll
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY 11549, USA..
| | - Henna Chaudhry
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY 11549, USA..
- Department of Chemistry, Hofstra University, Hempstead, NY 11549, USA
| | - Michelle Paszek
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY 11549, USA..
| | - Nancy Radecker
- Department of Biology, Hofstra University, Hempstead, NY 11549, USA
| | - Thomas Palaia
- Department of Foundations of Medicine, New York University School of Medicine, Mineola, NY 11501, USA
| | - Hazel Consunji de Guzman
- Department of Foundations of Medicine, New York University School of Medicine, Mineola, NY 11501, USA
- Hair Life Regeneration LLC, Copiague, NY 11726, USA
| | - Roche C de Guzman
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY 11549, USA..
| |
Collapse
|
27
|
Sharma S, Kishen A. Dysfunctional crosstalk between macrophages and fibroblasts under LPS-infected and hyperglycemic environment in diabetic wounds. Sci Rep 2025; 15:17233. [PMID: 40383800 PMCID: PMC12086240 DOI: 10.1038/s41598-025-00673-4] [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: 10/21/2024] [Accepted: 04/29/2025] [Indexed: 05/20/2025] Open
Abstract
Diabetic wounds, especially diabetic foot ulcers, present a major clinical challenge due to delayed healing and prolonged inflammation. Macrophage-fibroblast interactions are essential for wound repair, yet this crosstalk is disrupted in diabetic wounds due to hyperglycemia and bacterial infection. This study investigates the dysfunctional communication between macrophages and fibroblasts, focusing on autocrine, paracrine, and juxtacrine signaling in simulated diabetic environments. Using monoculture and co-culture models of THP-1-derived macrophages and primary human dermal fibroblasts, we simulated conditions of normal glucose, LPS-induced infection, high glucose (with AGEs), and combined high glucose (with AGEs) and LPS. Macrophages in hyperglycemic and LPS-infected environments exhibited a pro-inflammatory M1 phenotype with elevated expression of CD80, and STAT1 and increased production of IL-1β, TNF-α, and MMP9. Fibroblast migration was significantly impaired under high glucose conditions, particularly in paracrine model. Secretome profiling showed heightened pro-inflammatory cytokines and proteases, with reduced anti-inflammatory markers (IL-10 and VEGF-A) under hyperglycemic conditions. Paracrine signaling exacerbated the inflammatory response, while juxtacrine signaling showed more moderate effects, conducive to healing. These findings highlight the pathological macrophage-fibroblast crosstalk in diabetic wounds, particularly under hyperglycemic and LPS-infected conditions, offering insights for potential immunomodulatory therapies aimed at restoring effective signaling and improving wound healing outcomes.
Collapse
Affiliation(s)
- Shivam Sharma
- The Kishen Lab, Dental Research Institute, University of Toronto, Toronto, Canada
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON, M5G 1G6, Canada
| | - Anil Kishen
- The Kishen Lab, Dental Research Institute, University of Toronto, Toronto, Canada.
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, ON, M5G 1G6, Canada.
- Department of Dentistry, Mount Sinai Hospital, Toronto, Canada.
| |
Collapse
|
28
|
Shah S, D'Souza GGM. Modeling Tumor Microenvironment Complexity In Vitro: Spheroids as Physiologically Relevant Tumor Models and Strategies for Their Analysis. Cells 2025; 14:732. [PMID: 40422235 DOI: 10.3390/cells14100732] [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: 04/18/2025] [Revised: 05/11/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Drug delivery to solid tumors is challenged by multiple physiological barriers arising from the tumor microenvironment, including dense extracellular matrix, cellular heterogeneity, hypoxic gradients, and elevated interstitial fluid pressure. These features hinder the uniform distribution and accumulation of therapeutics, reducing treatment efficacy. Despite their widespread use, conventional two-dimensional monolayer cultures fail to reproduce these complexities, contributing to the poor translational predictability of many preclinical candidates. Three-dimensional multicellular tumor spheroids have emerged as more representative in vitro models that capture essential features of tumor architecture, stromal interactions, and microenvironmental resistance mechanisms. Spheroids exhibit spatially organized regions of proliferation, quiescence, and hypoxia, and can incorporate non-tumor cells to mimic tumor-stroma crosstalk. Advances in spheroid analysis now enable detailed evaluation of drug penetration, cellular migration, cytotoxic response, and molecular gradients using techniques such as optical and confocal imaging, large-particle flow cytometry, biochemical viability assays, and microfluidic integration. By combining physiological relevance with analytical accessibility, spheroid models support mechanistic studies of drug transport and efficacy under tumor-like conditions. Their adoption into routine preclinical workflows has the potential to improve translational accuracy while reducing reliance on animal models.
Collapse
Affiliation(s)
- Shrey Shah
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
- Atom Bioworks Inc., Cary, NC 27513, USA
| | - Gerard G M D'Souza
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
| |
Collapse
|
29
|
Kirchhoff R, Chromik MA, Schebb NH. Phagocytosis is differentially regulated by LPS in M1- and M2-like macrophages via PGE 2 formation and EP4 signaling. Prostaglandins Other Lipid Mediat 2025; 178:106998. [PMID: 40383415 DOI: 10.1016/j.prostaglandins.2025.106998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/30/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
Phagocytosis is a key process in human innate immune response. Human macrophages are important phagocytes engulfing and neutralizing pathogens and cell debris. In addition, they modulate the inflammatory process by releasing cytokines and lipid mediators. However, the link between oxylipins and phagocytosis in different macrophage phenotypes remains poorly understood. In order to better understand the link between phagocytosis and the arachidonic acid (ARA) cascade, we established a phagocytosis assay in primary human 'inflammatory' M1- and 'anti-inflammatory' M2-like macrophages from peripheral blood mononuclear cells (PBMC), representing extremes of macrophage phenotypes. The branches of the ARA cascade were investigated by quantitative targeted proteomics and metabolomics. M1-like macrophages show a higher abundance of cyclooxygenase (COX)-2 and its products particularly after LPS stimulus compared to M2-like macrophages. LPS increased phagocytosis in M2-like, but not in M1-like macrophages. We demonstrate that the COX product prostaglandin E2 (PGE2) modulates the differential effects of LPS on phagocytosis: Via the EP4 receptor PGE2 signaling suppresses phagocytosis in primary human macrophages. Thus, blockage of COX, e.g. by non-steroidal anti-inflammatory drugs (NSAID), leads to an increase of phagocytosis also in 'inflammatory' M1-like macrophages. This supports the well-described anti-inflammatory effects of these drugs and underscores the importance of the link between the COX branch of the ARA cascade and the regulation of phagocytosis in human macrophages.
Collapse
Affiliation(s)
- Rebecca Kirchhoff
- Chair of Food Chemistry, School of Mathematics and Natural Sciences, University of Wuppertal, Gaussstr. 20, Wuppertal 42119, Germany
| | - Michel André Chromik
- Chair of Food Chemistry, School of Mathematics and Natural Sciences, University of Wuppertal, Gaussstr. 20, Wuppertal 42119, Germany
| | - Nils Helge Schebb
- Chair of Food Chemistry, School of Mathematics and Natural Sciences, University of Wuppertal, Gaussstr. 20, Wuppertal 42119, Germany.
| |
Collapse
|
30
|
Wu T, Wang Y, Xia Y, Ji J, Tao X, Gu Z. Identifying the genetic association between rheumatoid arthritis and the risk of infectious diseases. Clin Rheumatol 2025:10.1007/s10067-025-07485-x. [PMID: 40377766 DOI: 10.1007/s10067-025-07485-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 12/15/2024] [Accepted: 05/05/2025] [Indexed: 05/18/2025]
Abstract
BACKGROUND Previous evidence suggests an association between rheumatoid arthritis (RA) and infectious diseases, but the causal relationship remains unclear. This study sought to explore causal associations between RA and five common infections: pneumonia, sepsis, urinary tract infections (UTI), skin and subcutaneous tissue infections (SSTI), and bacterial intestinal infections (BII). METHODS To identify the causal links, we adopted a Mendelian randomization (MR) design utilizing the inverse variance weighted (IVW), weighted median (WM), and MR-Egger approaches. Univariable MR (UVMR) and multivariable MR (MVMR) analyses were performed using pooled genome-wide association studies (GWAS) data. Additionally, various sensitivity analyses were conducted to ensure the reliability of the results. RESULTS In the UVMR analysis, RA was potentially associated with elevated risks of pneumonia (OR = 1.034, 95% CI: 1.016-1.052, P < 0.001) and sepsis (OR = 1.079, 95% CI: 1.048-1.110, P = 3.507E-07). This association remained significant after adjusting for smoking, alcohol consumption, or type 2 diabetes mellitus (T2DM) in the MVMR analysis. However, no causal links were found between RA and UTI, SSTI, and BII. Sensitivity analyses showed no detectable heterogeneity or pleiotropy, strengthening the causal inference of results. CONCLUSION Our study provides strong evidence of the association between RA and increased risks of pneumonia and sepsis. Further research is required to validate these findings and elucidate the underlying mechanisms. Key Points • It remains unclear whether the increased susceptibility to infections in RA stems from a genetic predisposition or results from immunosuppressive treatments. • The MR method is employed to assess the relationship between RA and common infectious diseases. • Our MR study supports a potential causal relationship between RA and elevated risks of pneumonia and sepsis.
Collapse
Affiliation(s)
- Teng Wu
- Dalian Medical University, Dalian, China
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yunan Wang
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yunfei Xia
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
| | - Juan Ji
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xinyu Tao
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
| | - Zhifeng Gu
- Dalian Medical University, Dalian, China.
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China.
| |
Collapse
|
31
|
Lund A, Moffat D, Jepsen SD, Desler C, Andresen L, Bregenholt S, Reddy V, Skov S. Targeted p38 mitogen-activated protein kinase inhibition potently augment inflammatory responses of human macrophages toward Staphylococcus aureus. Mol Immunol 2025; 183:145-155. [PMID: 40373637 DOI: 10.1016/j.molimm.2025.05.010] [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: 03/14/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Antibiotic-resistant Staphylococcus aureus (S. aureus) is a growing challenge for human health and novel treatment options are needed. Here we examine a novel therapeutic approach against persistent S. aureus infections based on monocyte/macrophage specific inhibition of the p38α mitogen-activated protein kinase activity. Since systemic p38α kinase inhibition cause aberrant toxicity, we used the myeloid specific p38α kinase inhibitor, MPL-5821. P38α kinase inhibition caused a potent increase in the pro-inflammatory profile of human macrophages after exposure to S. aureus, including upregulation of M1-markers and induction of pro-inflammatory cytokines including IFN-γ, TNF-α, IL-1β, IL12p70, IL-6 and IL-8, as well as an increase in phagocytic capacities. These pro-inflammatory signals were only seen after combined S. aureus exposure and p38α inhibition. Macrophages are often regulated by changes in intracellular metabolism. In agreement with this, the combination of S. aureus exposure and p38α inhibition led to specific changes in glycolytic and mitochondrial activity within the responding macrophages. Our study thus unravels a novel and specific activation of macrophages that augment their response toward S. aureus, without causing aberrant inflammation. This constitutes a unique non-antibiotic therapeutic approach that can potentially be used against persistent S. aureus infection.
Collapse
Affiliation(s)
- Astrid Lund
- Laboratory of Immunology, Section for Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - David Moffat
- Macrophage Pharma, 59-60 Thames St, Windsor SL4 1TX, United Kingdom
| | - Stine Dam Jepsen
- Laboratory of Immunology, Section for Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Claus Desler
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Andresen
- Laboratory of Immunology, Section for Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Søren Bregenholt
- Macrophage Pharma, 59-60 Thames St, Windsor SL4 1TX, United Kingdom
| | - Venkat Reddy
- Macrophage Pharma, 59-60 Thames St, Windsor SL4 1TX, United Kingdom
| | - Søren Skov
- Laboratory of Immunology, Section for Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
32
|
Sutera S, Furchì OA, Pentenero M. Macrophages and the immune microenvironment in OPMDs: a systematic review of the literature. FRONTIERS IN ORAL HEALTH 2025; 6:1605978. [PMID: 40432828 PMCID: PMC12106459 DOI: 10.3389/froh.2025.1605978] [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: 04/04/2025] [Accepted: 04/17/2025] [Indexed: 05/29/2025] Open
Abstract
Background In the presence of cancers, Tumor Associated Macrophages have a well-established role, but the literature provides limited evidence regarding their involvement in the onset and malignant transformation of Oral Potentially Malignant Disorders (OPMDs). Objectives The present systematic review aimed to collect evidence on the presence and characterization of macrophages in the microenvironment of OPMDs. Data sources PubMed, Scopus, EMBASE, Web of Science. Study eligibility criteria Ex vivo or in silico human studies reporting original quantitative data on macrophage infiltration in OPMDs or Oral Epithelial Dysplasia (OED), published from 1990 onward. Results Thirty-seven studies were included for qualitative analysis. Investigated OPMDs included: oral leukoplakia, oral lichen planus, oral lichenoid lesions, proliferative leukoplakia, oral submucous fibrosis, actinic cheilitis, chronic graft vs. host disease. Discussion Even though the heterogeneity of data from the included studies prevents a meta-analysis, the reported results are quite consistent in supporting an increasing macrophage infiltration from normal mucosa to OPMDs, OED, and Oral Squamous Cell Carcinoma (OSCC). An M1 pro-inflammatory polarization is prevalent in OPMDs, with a shift toward an M2 pro-tumorigenic polarization in moderate-severe OED and OSCC. Several novel markers including STAT1, IDO, PD-L1, APOE, ITGB2 appear to be able to identify macrophage clusters involved in pro-inflammatory or pro-tumorigenic pathways. Conclusions Evidence from the present review supports an active role of macrophages in regulating immune suppression, oncogenesis, and tumor progression in OPMDs and during the transition to OSCC. Future research should focus not merely on cell quantification and general M1/M2 polarization but rather on the expression of specific markers potentially linked to immunomodulatory pathways involved in oncogenesis.
Collapse
|
33
|
Feng W, Yang K, Zou Y, Xiao Z, Qian R, Qian R. Progress of ursolic acid on the regulation of macrophage: summary and prospect. Front Immunol 2025; 16:1576771. [PMID: 40421013 PMCID: PMC12104263 DOI: 10.3389/fimmu.2025.1576771] [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: 02/14/2025] [Accepted: 04/15/2025] [Indexed: 05/28/2025] Open
Abstract
Ursolic acid (UA), a prevalent pentacyclic triterpenoid found in numerous fruits and herbs, has garnered significant attention for its vital role in anti-inflammatory processes and immune regulation. The study of immune cells has consistently been a focal point, particularly regarding macrophages, which play crucial roles in antigen presentation, immunomodulation, the inflammatory response, and pathogen phagocytosis. This paper reveals the underlying regulatory effects of UA on the function of macrophages and the specific therapeutic effects of UA on a variety of diseases. Owing to the superior effect of UA on macrophages, different types of macrophages in different tissues have been described. Through the multifaceted regulation of macrophage function, UA may provide new ideas for the development of novel anti-inflammatory and immunomodulatory drugs. However, to facilitate its translation into actual medical means, the specific mechanism of UA in macrophages and its clinical application still need to be further studied.
Collapse
Affiliation(s)
- Wenjing Feng
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Kehong Yang
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Ying Zou
- Department of Anatomy, Anatomy Teaching Center of Hunan University of Chinese Medicine, Changsha, China
| | - Zhaohua Xiao
- Xiangya Hospital, Central South University, Changsha, China
| | - Rongkang Qian
- Department of Integrated Traditional Chinese and Western Medicine, Qian Rongkang Clinic, Loudi, China
| | - Ronghua Qian
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
34
|
Pastrana-Otero I, Godbole AR, Kraft ML. Noninvasive and in situ identification of the phenotypes and differentiation stages of individual living cells entrapped within hydrogels. Analyst 2025; 150:2047-2057. [PMID: 40198151 PMCID: PMC11977708 DOI: 10.1039/d4an00800f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 03/29/2025] [Indexed: 04/10/2025]
Abstract
Microscale screening platforms that allow cells to interact in three dimensions (3D) with their microenviroment have been developed as a tool for identifying the extrinsic cues that might stimulate stem cells to replicate without differentiating within artificial cultures. Though these platforms reduce the number of valuable stem cells that must be used for screening, analyzing the fate decisions of cells in these platforms can be challenging. New noninvasive approaches for identifying the lineage-specific differentiation stages of cells while they are entrapped in the hydrogels used for these 3D cultures are especially needed. Here we used Raman spectra acquired from individual, living cells entrapped within a hydrogel matrix and multivariate analysis to identify cell phenotype noninvasively and in situ. We collected a single Raman spectrum from each cell of interest while it was entrapped within a hydrogel matrix and used partial least-squares discriminant analysis (PLS-DA) of the spectra for cell phenotype identification. We first demonstrate that this approach enables identifying the lineages of individual, living cells from different laboratory lines entrapped within two different hydrogels that are used for 3D culture, collagen and gelatin methacrylate (gelMA). Then we use a hematopoietic progenitor cell line that differentiates into different types of macrophages to show that the lineage-specific differentiation stages of individual, living hematopoietic cells entrapped inside of gelMA scaffolds may be identified by PLS-DA of Raman spectra. This ability to noninvasively identify the lineage-specific differentiation stages of cells without removing them from a 3D culture could enable tracking the differentiation of the same cell over time.
Collapse
Affiliation(s)
- Isamar Pastrana-Otero
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Apurva R Godbole
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Mary L Kraft
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, USA.
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, USA
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, USA
| |
Collapse
|
35
|
Liu B, Peng Y, Wang C, Wei H, Xu S, Liu Y, Yin X, Bi H, Guo D. Baicalin prevents experimental autoimmune uveitis by promoting macrophage polarization balance through inhibiting the HIF-1α signaling pathway. Sci Rep 2025; 15:16424. [PMID: 40355594 PMCID: PMC12069706 DOI: 10.1038/s41598-025-01286-7] [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: 02/24/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
Uveitis is a series of autoimmune eye diseases that can seriously damage people's eyesight. This study aimed to explore the therapeutic potential of baicalin in treating uveitis, focusing on its modulation of HIF-1α expression and macrophage polarization. Using an experimental autoimmune uveitis (EAU) rat model, we found that baicalin can significantly reduce fundus inflammation in EAU rats. Spectral-domain optical coherence tomography revealed retinal vascular thickening in the EAU group, indicating severe inflammation, which baicalin effectively mitigated. Histopathological analysis confirmed reduced inflammatory cell infiltration in the ciliary body and retina. Co-immunoprecipitation analyses showed that HIF-1αinteracted with macrophage-related factors, including iNOS and ARG1. Baicalin downregulated HIF-1α and iNOS while upregulating ARG1, balancing pro-inflammatory M1 and anti-inflammatory M2 macrophage polarization. Flow cytometry demonstrated a reversal of M1/M2 macrophage ratios in the EAU group after baicalin treatment. Additionally, baicalin improved macrophage mitochondrial membrane potential and decreased reactive oxygen species (ROS) levels, shifting macrophage polarization toward an anti-inflammatory state. These findings confirm that baicalin can effectively reduce inflammation and restore immune balance by orchestrating the HIF-1α pathway, establishing a promising therapeutic candidate for uveitis and highlighting the potential of natural bioactive compounds in treating and preventing inflammatory diseases through targeted immune modulation.
Collapse
Affiliation(s)
- Bin Liu
- Shandong University of Traditional Chinese Medicine, No. 4655#, Daxue Road, Jinan, 250355, China
| | - Yuan Peng
- Shandong University of Traditional Chinese Medicine, No. 4655#, Daxue Road, Jinan, 250355, China
| | - Congling Wang
- Shandong University of Traditional Chinese Medicine, No. 4655#, Daxue Road, Jinan, 250355, China
| | - Huixia Wei
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
- Shandong Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, 250002, China
| | - Shuqin Xu
- Shandong University of Traditional Chinese Medicine, No. 4655#, Daxue Road, Jinan, 250355, China
| | - Yunfeng Liu
- Shandong University of Traditional Chinese Medicine, No. 4655#, Daxue Road, Jinan, 250355, China
| | - Xuewei Yin
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
- Shandong Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, 250002, China
| | - Hongsheng Bi
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
- Shandong Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, 250002, China
| | - Dadong Guo
- Shandong Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, 250002, China.
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, 250002, China.
- Shandong Academy of Eye Disease Prevention and Therapy, No. 48#, Yingxiongshan Road, Jinan, 250002, China.
| |
Collapse
|
36
|
An T, Guo M, Wang Z, Liu K. Tissue-Resident Macrophages in Cardiovascular Diseases: Heterogeneity and Therapeutic Potential. Int J Mol Sci 2025; 26:4524. [PMID: 40429668 PMCID: PMC12111180 DOI: 10.3390/ijms26104524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Tissue-resident macrophages (TRMs) play a crucial role in maintaining tissue homeostasis and regulating immune responses. In recent years, an increasing number of studies have highlighted their central role in cardiovascular diseases. This review provides a comprehensive overview of TRMs, with a particular emphasis on cardiac resident macrophages (CRMs), discussing their origin, heterogeneity, and functions in various cardiovascular diseases. We conduct an in-depth analysis of macrophage subpopulations based on C-C Chemokine Receptor Type 2 (CCR2) receptor expression, elucidating the role of CCR2+ macrophages in promoting fibrosis and cardiac remodeling, while highlighting the protective functions of CCR2- macrophages in suppressing inflammation and promoting tissue repair. In atherosclerosis, we focus on the role of metabolic reprogramming in regulating macrophage polarization, revealing how metabolic pathways influence the balance between pro-inflammatory M1 and anti-inflammatory M2 macrophages, thereby affecting plaque stability and disease progression. By summarizing the roles of these macrophage subpopulations in myocardial infarction, heart failure, and other diseases, we propose potential therapeutic strategies aimed at modulating different macrophage subtypes. These include targeting the CCR2 signaling pathway to mitigate inflammation and fibrosis, and metabolic reprogramming to restore the balance between M1 and M2 macrophages. Finally, we highlight the need for future research to focus on the functional diversity and molecular mechanisms of human TRMs to develop novel immunotherapeutic strategies and improve the prognosis of cardiovascular diseases.
Collapse
Affiliation(s)
- Tianhui An
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Mengyuan Guo
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Zhaohui Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Kun Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| |
Collapse
|
37
|
Zheng Z, Lin L, Lin H, Zhou J, Wang Z, Wang Y, Chen J, Lai C, Li R, Shen Z, Zhong M, Xie C, Chen Y, Zhang X, Guo Z, Dong R, He S, Chen F. Acetylcholine from tuft cells promotes M2 macrophages polarization in Hirschsprung-associated enterocolitis. Front Immunol 2025; 16:1559966. [PMID: 40416975 PMCID: PMC12098611 DOI: 10.3389/fimmu.2025.1559966] [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: 01/13/2025] [Accepted: 04/08/2025] [Indexed: 05/27/2025] Open
Abstract
Background Hirschsprung-associated enterocolitis (HAEC) is one of the most severe complications in patients with Hirschsprung's disease (HSCR). Previous research has indicated that acetylcholine (ACH) plays an anti-inflammatory role during inflammation by acting on the α7 nicotinic acetylcholine receptor(α7nAchR) to promote the secretion of anti-inflammatory factors. However, the specific role of ACH in HAEC remains unclear. This experiment aims to explore the sources of ACH in HSCR and its anti-inflammatory mechanisms, thereby identifying new directions for the prevention and treatment of HAEC. Methods We analyzed single-cell transcriptome data from HSCR to identify cells that secrete ACH and observed their distribution using immunofluorescence. In Ednrb-/- mice, F4/80, iNOS, ARG-1 and CD206 were used to identify and locate M1 and M2 macrophages in different intestinal segments. Western blot, reverse transcription-quantitative polymerase chain reaction, and enzyme-linked immunosorbent assay were used to test the levels of IκBα, tumor necrosis factor-α, interleukin-10, and the macrophage activation pathway proteins JAK2 and STAT3 in different intestinal segments of Ednrb-/- mice. Organoid and cell culture techniques were used to verify the anti-inflammatory mechanism of ACH in vitro models. Results scRNA-seq analysis revealed that tuft cells expressed the CHAT protein. In HSCR, aganglionic segments exhibited heightened cholinergic activity compared with dilated ganglionic segments. In HAEC, inflammation was mainly concentrated in the dilated ganglionic segment and was associated with an increase in M1 macrophages, whereas the aganglionic segment showed less inflammation and was associated with an increase in M2 macrophages. Furthermore, in vitro experiments showed that intestinal organoids containing tuft cells promoted an increase in M2 macrophage markers, and ACH promoted M2 macrophage polarization. Conclusions Differences in inflammation among various intestinal segments in HAEC may be linked to ACH secreted by tuft cells. Drugs targeting tuft cells have the potential to become important components of HAEC treatment in the future.
Collapse
Affiliation(s)
- Ziyi Zheng
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lin Lin
- Institute of Population Medicine, School of Public Health, Fujian Medical University, University Town, Fuzhou, China
| | - Huifang Lin
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jie Zhou
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhe Wang
- Institute of Population Medicine, School of Public Health, Fujian Medical University, University Town, Fuzhou, China
| | - Yang Wang
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jianxin Chen
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Caimin Lai
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Renfu Li
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhiyong Shen
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ming Zhong
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Cheng Xie
- Fuzhou Children’s Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Yinjian Chen
- Fujian Children’s Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Xuechao Zhang
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhongjie Guo
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Rui Dong
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
- Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Shiwei He
- Institute of Population Medicine, School of Public Health, Fujian Medical University, University Town, Fuzhou, China
| | - Feng Chen
- Department of Pediatric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
38
|
Ivy A, Bess SN, Agrawal S, Kochar V, Stokes AL, Muldoon TJ, Nelson CE. A dual-fluorescence assay for gene delivery vehicle screening in macrophages with an inflammation-inducible reporter construct. BMC METHODS 2025; 2:8. [PMID: 40352095 PMCID: PMC12062070 DOI: 10.1186/s44330-025-00030-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
Background Macrophages are a promising target for therapeutics in various applications such as regenerative medicine and immunotherapy for cancer. Due to their plastic nature, macrophages can switch from a non-activated state to activated with the smallest environmental change. For macrophages to be effective in their respective applications, screening for phenotypic changes is necessary to elucidate the cell response to different delivery vehicles, vaccines, small molecules, and other stimuli. Methods We created a sensitive and dynamic high-throughput screening method for macrophages based on the activation of NF-κB. For this reporter, we placed an mRFP1 fluorescence gene under the control of an inflammatory promoter, which recruits NF-κB response elements to promote expression during the inflammatory response in macrophages. We characterized the inflammatory reporter based on key markers of an inflammatory response in macrophages including TNF-α cytokine release and immunostaining for inflammatory and non-inflammatory cell surface markers. We compared gene delivery and inflammation of several clinically relevant viral vehicles and commercially available non-viral vehicles. Statistical analysis between groups was performed with a one-way ANOVA with post-hoc Tukey's test. Results The reporter macrophages demonstrated a dynamic range after LPS stimulation with an EC50 of 0.61 ng/mL that was highly predictive of TNF-α release. Flow cytometry revealed heterogeneity between groups but confirmed population level shifts in pro-inflammatory markers. Finally, we demonstrated utility of the reporter by showing divergent effects with various leading gene delivery vehicles. Discussion This screening technique developed here provides a dynamic, high-throughput screening technique for determining inflammatory response by mouse macrophages to specific stimuli. The method presented here provides insight into the inflammatory response in mouse macrophages to different viral and non-viral gene delivery methods and provides a tool for high-throughput screening of novel vehicles. Supplementary Information The online version contains supplementary material available at 10.1186/s44330-025-00030-x.
Collapse
Affiliation(s)
- Allie Ivy
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Shelby N. Bess
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Shilpi Agrawal
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Varun Kochar
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Abbey L. Stokes
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
| | - Timothy J. Muldoon
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR USA
| | - Christopher E. Nelson
- Department of Biomedical Engineering, University of Arkansas, 120 John A. White Jr. Engineering Hall, Fayetteville, AR 72701 USA
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR USA
| |
Collapse
|
39
|
Jiang M, Bu W, Wang X, Ruan J, Shi W, Yu S, Huang L, Xue P, Tang J, Zhao X, Su L, Cheng D. Pulmonary fibrosis: from mechanisms to therapies. J Transl Med 2025; 23:515. [PMID: 40340941 PMCID: PMC12063347 DOI: 10.1186/s12967-025-06514-2] [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: 02/07/2025] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive interstitial lung disease characterized by excessive deposition of extracellular matrix (ECM) and abnormal fibroblast proliferation, which is mainly caused by air pollution, smoking, aging, occupational exposure, environmental pollutants exposure, and microbial infections. Although antifibrotic agents such as pirfenidone and nintedanib, approved by the United States (US) Food and Drug Administration (FDA), can slow the decline in lung function and disease progression, their side effects and delivery inefficiency limit the overall prognosis of PF. Therefore, there is an urgent need to develop effective therapeutic targets and delivery approaches for PF in clinical settings. This review provides an overview of the pathogenic mechanisms, therapeutic drug targeting signaling pathways, and promising drug delivery strategies for treating PF.
Collapse
Affiliation(s)
- Mengna Jiang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Wenxia Bu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Xuehai Wang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Jialing Ruan
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Weijian Shi
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Siqi Yu
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, 334000, China
| | - Lizhen Huang
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, 334000, China
| | - Peng Xue
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Juan Tang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China.
| | - Liling Su
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, 334000, China.
| | - Demin Cheng
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China.
| |
Collapse
|
40
|
Cuijpers I, Katsburg J, van Loon LJC, Troost FJ, Sthijns MMJPE. Nutritional strategies targeting age-related skeletal muscle fibrosis: underlying mechanisms. Crit Rev Food Sci Nutr 2025:1-21. [PMID: 40336331 DOI: 10.1080/10408398.2025.2498676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Aging is associated with a reduced number and function of muscle stem cells (MuSC). This results in a decreased muscle regenerative capacity and increased formation of fibrotic tissue, impairing skeletal muscle function. This review provides an overview of in vitro and in vivo animal studies investigating nutritional interventions with the potential to inhibit pathophysiological mechanisms involved in the development of skeletal muscle fibrosis. Mechanism targets include 1) MuSC function and myogenic differentiation, 2) M1 to M2 macrophage polarization, 3) myofibroblast activity or extracellular matrix (ECM) deposition, and 4) reactive oxygen species (ROS) mediated pathways, such as NOX2/4 activity. Most promising nutrients described in this review are phytonutrients, vitamins and amino acids. Quercetin targets multiple pathways (showing decreased inflammation, ECM expression and NOX2/4 activity) in various cell types and tissues (kidney, aorta, liver and (heart) muscle) of rodents and rabbits, which could contribute to fibrosis development. Additionally, sulforaphane is a promising candidate as it inhibits inflammation, ECM expression, and ROS production in mouse skeletal muscle. After validation of the effects in human skeletal muscle, supplementation with these nutrients could be implemented in a multifaceted intervention (including exercise and adequate protein intake) targeting age-related skeletal muscle fibrosis.
Collapse
Affiliation(s)
- Iris Cuijpers
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Joey Katsburg
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Luc J C van Loon
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Freddy J Troost
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Mireille M J P E Sthijns
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| |
Collapse
|
41
|
Sun L, Gao X, Zhao Z, Zhu Y. Molecular subtypes and quantitative analysis of PD-L1 and tumor-associated immune cells in uterine carcinosarcoma. J Gynecol Oncol 2025; 36:36.e114. [PMID: 40405429 DOI: 10.3802/jgo.2025.36.e114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/24/2025] [Accepted: 04/03/2025] [Indexed: 05/24/2025] Open
Abstract
OBJECTIVE In the present study, molecular subtypes were determined, programmed death-ligand 1 (PD-L1) and tumor-associated immune cells (TAICs) were quantitatively detected, and their effect on prognosis in uterine carcinosarcoma (UCS) was analyzed. METHODS The study included 65 UCS cases. Direct sequencing of POLE exonuclease domain and immunohistochemistry of mismatch repair (MMR) deficiency proteins and p53 were used to stratify molecular subtypes. QuPath was used for quantitative immunohistochemical detection of PD-L1 and TAICs. The chi square test was used to determine the association between molecular subtypes and expression of PD-L1 and TAICs. The Kaplan-Meier method and Cox proportional hazards regression were used for plotting and survival analysis. RESULTS In 65 UCS cases, 1 case (1.5%) was POLE ultramutated (POLEmut) subtype, 11 cases (16.9%) were deficient MMR (dMMR) subtype, 32 cases (49.3%) were p53 mutant (p53mut) subtype, and 21 cases (32.3%) were nonspecific molecular profile (NSMP) subtype. The positive density of PD-L1 in tumor (p=0.022), CD8 in stroma (p=0.036), and CD163 in stroma (p=0.025) were significantly associated with molecular subtypes. The patients with POLEmut and dMMR subtypes had a relatively better prognosis trend than patients with NSMP and p53mut subtypes. The patients with high positive density of PD-L1 in tumor had significantly better prognosis; however, high positive density of CD163 in stroma showed significantly worse prognosis. CONCLUSION UCS could be classified into four molecular subtypes associated with prognosis. PD-L1 and M2 macrophages could effectively predict the prognosis of patients with UCS.
Collapse
Affiliation(s)
- Lili Sun
- Department of Pathology, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Xiaozhuo Gao
- Department of Pathology, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Zehua Zhao
- Department of Pathology, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Yanmei Zhu
- Department of Pathology, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China.
| |
Collapse
|
42
|
Li NY, Vorrius B, Rebello E, Ge J, Mohite A, Qiao Z, Ding J, Chen Q. Matrilin-2 with a K-Chitosan Scaffold Enhances Functional Recovery and Nerve Regeneration in a Segmental Rat Sciatic Nerve Injury Model. Pharmaceuticals (Basel) 2025; 18:686. [PMID: 40430505 PMCID: PMC12114659 DOI: 10.3390/ph18050686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Previous work in our lab demonstrated that a 3D scaffold containing lysine-modified chitosan (K-chitosan) and decorated with Matrilin-2 (MATN2) enhanced Schwann cell (SC) migration and axonal outgrowth in vitro and ex vivo. This study aimed to assess the regenerative effect of this scaffold compared to that of a collagen conduit and an autograft using a segmental rat sciatic nerve injury model. Methods: A total of 30 Lewis Rats were assigned into three groups: an untreated collagen conduit (UC) group, a collagen conduit treated with MATN2 K-chitosan (TC) group, and a reverse autograft (RA) group. Walking force measurements, compound muscle action potential (CMAP), the wet muscle weight of the tibialis anterior and the gastrocnemius, and axonal histomorphometry were assessed. Results: The walking force and CMAP were significantly higher in the TC group compared to those in the UC group, with no significant difference between the TC and RA groups. The muscle weights were significantly greater in the TC group compared to those in the UC group but smaller than those in the RA group. The TC group experienced significantly greater axonal regeneration compared to that with the UC, and no differences were found with the RA. The TC group further demonstrated significantly greater cell counts than those in the UC group and greater affinity of the Schwann cells towards nerve reconstruction. Conclusion: The MATN2 K-chitosan scaffold significantly improved nerve regeneration and was comparable to the RA, supporting the development of a novel bio-conductive scaffold conduit.
Collapse
Affiliation(s)
- Neill Y. Li
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Brown University Health, Providence, RI 02912, USA; (B.V.); (E.R.); (J.G.); (J.D.)
- Laboratory for Peripheral Neuroregenerative Biotherapeutics, Duke Nerve Center, Durham, NC 27710, USA;
- Division of Hand, Upper Extremity, and Microvascular Surgery, Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Brandon Vorrius
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Brown University Health, Providence, RI 02912, USA; (B.V.); (E.R.); (J.G.); (J.D.)
| | - Elliott Rebello
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Brown University Health, Providence, RI 02912, USA; (B.V.); (E.R.); (J.G.); (J.D.)
| | - Jonathan Ge
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Brown University Health, Providence, RI 02912, USA; (B.V.); (E.R.); (J.G.); (J.D.)
| | - Amit Mohite
- Laboratory for Peripheral Neuroregenerative Biotherapeutics, Duke Nerve Center, Durham, NC 27710, USA;
| | - Zhen Qiao
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Brown University Health, Providence, RI 02912, USA; (B.V.); (E.R.); (J.G.); (J.D.)
| | - Jing Ding
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Brown University Health, Providence, RI 02912, USA; (B.V.); (E.R.); (J.G.); (J.D.)
| | - Qian Chen
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Brown University Health, Providence, RI 02912, USA; (B.V.); (E.R.); (J.G.); (J.D.)
| |
Collapse
|
43
|
Wang Y, Gao J, Wu T, Wang Z. M2 Macrophages Mitigate Ocular Surface Inflammation and Promote Recovery in a Mouse Model of Dry Eye. Ocul Immunol Inflamm 2025:1-10. [PMID: 40327794 DOI: 10.1080/09273948.2025.2497484] [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: 03/27/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025]
Abstract
PURPOSE Dry eye disease (DED) is a chronic, progressive, multifactorial condition characterized by tear film instability and ocular surface damage. Ocular surface inflammation is one of the main mechanisms of DED. This study aims to investigate the therapeutic effects of anti-inflammatory M2 macrophages on ocular surface inflammation and their potential mechanisms in improving dry eye symptoms in a mouse model. METHODS Mouse macrophages (RAW264.7) were polarized into M2 macrophages by IL-4 under different osmolarities, and M2 macrophage conditioned medium (M2-CM) was collected. Flow cytometry and ELISA were applied to measure the cytokine expression of the M2 macrophages. Primary mouse corneal epithelial cells (CECs) were co-cultured with RAW264.7 and M2 macrophages using a Transwell system. The viability and migration of CECs were assessed using CCK-8 and scratch assays. Mouse DED was established by subcutaneous injection of scopolamine, and the therapeutic effects of M2-CM were evaluated by phenol red thread test, fluorescein staining, and tear film breakup time (BUT). PCR and immunofluorescence staining were applied to observe inflammatory factors and cells on the ocular surface. RESULTS M2 macrophages enhanced CEC viability, proliferation, and migration, but hyperosmolarity inhibited M2 macrophage polarization. In the DED model, M2-CM improved ocular surface conditions, reduced pro-inflammatory cytokine expression, and increased anti-inflammatory factors. Immunofluorescence revealed reduced pro-inflammatory cells (M1 macrophages, Th1, and Th17) and increased M2 macrophages in the ocular tissues after M2-CM treatment. CONCLUSION These results suggest that M2-CM ameliorates ocular surface inflammation and promotes recovery in DED, offering a potential therapeutic strategy for DED.
Collapse
Affiliation(s)
- Yingming Wang
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jing Gao
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Tianhong Wu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhenyu Wang
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
44
|
Jiang H, Pang J, Li T, Akofala A, Zhou X, Yi C, Ning S, Gao X, Qiao Y, Kou J. PD-1 regulates the anti-tumor immune function of macrophages through JAK2-STAT3 signaling pathway in colorectal cancer tumor microenvironment. J Transl Med 2025; 23:502. [PMID: 40317043 PMCID: PMC12048993 DOI: 10.1186/s12967-025-06469-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/07/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs), as key immune components of the TME, play a pivotal role in tumor progression by fostering an immunosuppressive environment. Programmed death 1 (PD-1), a critical immune checkpoint molecule predominantly expressed on T cells, mediates immune suppression by binding to programmed death-ligand 1 (PD-L1) on tumor cells within the tumor microenvironment (TME). Emerging evidence reveals that TAMs also express PD-1, however, the expression and functional regulatory mechanisms of PD-1 on TAM remain poorly understood. METHODS In this study, we combined bulk RNA sequencing and single-cell RNA sequencing (scRNA-seq) data to investigate the association between PD-1 expression on macrophages and patient prognosis, while also uncovering the molecular mechanisms by which PD-1 regulates macrophage function. To further investigate the role of PD-1 in macrophage activity, we established a fluorescence-labeled tumor-bearing mouse model using CT26 cells, a murine colorectal cancer cell line, to evaluate the relationship between PD-1 expression on TAMs and their phagocytic activity as well as other functions. Additionally, to mimic the TME in vitro, we cultured bone marrow-derived macrophages (BMDMs) with CT26-conditioned medium (CT26-CM). RESULTS Our results suggest that PD-1 expression on TAMs drives macrophage polarization toward an M2-like phenotype, suppresses their phagocytic activity, inhibits the synthesis of interferon-γ (IFN-γ) signaling molecules, and ultimately promotes tumor progression. Mechanistically, we demonstrated that PD-1 regulates the synthesis of IFN-γ signaling molecules and the polarization of M2-type macrophages in BMDMs through the JAK2-STAT3 signaling pathway. Overall, our study demonstrates that PD-1 expression on TAMs facilitates the formation of an immunosuppressive microenvironment, ultimately accelerating tumor progression. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Han Jiang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, China
- Department of BioinformaticsScience and Technology, Harbin Medical University, Harbin, 150000, China
| | - Jingjing Pang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, China
| | - Tengyue Li
- Department of BioinformaticsScience and Technology, Harbin Medical University, Harbin, 150000, China
| | - Atitso Akofala
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, China
| | - Xiaoxi Zhou
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Changhua Yi
- The Second Hospital of Nanjing, Nanjing, 210003, China
| | - Shangwei Ning
- Department of BioinformaticsScience and Technology, Harbin Medical University, Harbin, 150000, China.
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, China.
- Basic Medical Institute of Heilongjiang Medical Sciences Academy, Harbin, 150086, China.
| | - Yu Qiao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, China.
| | - Jiayuan Kou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, China.
| |
Collapse
|
45
|
Zheng Y, Ren Z, Liu Y, Yan J, Chen C, He Y, Shi Y, Cheng F, Wang Q, Li C, Wang X. T cell interactions with microglia in immune-inflammatory processes of ischemic stroke. Neural Regen Res 2025; 20:1277-1292. [PMID: 39075894 PMCID: PMC11624874 DOI: 10.4103/nrr.nrr-d-23-01385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 01/17/2024] [Accepted: 03/07/2024] [Indexed: 07/31/2024] Open
Abstract
The primary mechanism of secondary injury after cerebral ischemia may be the brain inflammation that emerges after an ischemic stroke, which promotes neuronal death and inhibits nerve tissue regeneration. As the first immune cells to be activated after an ischemic stroke, microglia play an important immunomodulatory role in the progression of the condition. After an ischemic stroke, peripheral blood immune cells (mainly T cells) are recruited to the central nervous system by chemokines secreted by immune cells in the brain, where they interact with central nervous system cells (mainly microglia) to trigger a secondary neuroimmune response. This review summarizes the interactions between T cells and microglia in the immune-inflammatory processes of ischemic stroke. We found that, during ischemic stroke, T cells and microglia demonstrate a more pronounced synergistic effect. Th1, Th17, and M1 microglia can co-secrete pro-inflammatory factors, such as interferon-γ, tumor necrosis factor-α, and interleukin-1β, to promote neuroinflammation and exacerbate brain injury. Th2, Treg, and M2 microglia jointly secrete anti-inflammatory factors, such as interleukin-4, interleukin-10, and transforming growth factor-β, to inhibit the progression of neuroinflammation, as well as growth factors such as brain-derived neurotrophic factor to promote nerve regeneration and repair brain injury. Immune interactions between microglia and T cells influence the direction of the subsequent neuroinflammation, which in turn determines the prognosis of ischemic stroke patients. Clinical trials have been conducted on the ways to modulate the interactions between T cells and microglia toward anti-inflammatory communication using the immunosuppressant fingolimod or overdosing with Treg cells to promote neural tissue repair and reduce the damage caused by ischemic stroke. However, such studies have been relatively infrequent, and clinical experience is still insufficient. In summary, in ischemic stroke, T cell subsets and activated microglia act synergistically to regulate inflammatory progression, mainly by secreting inflammatory factors. In the future, a key research direction for ischemic stroke treatment could be rooted in the enhancement of anti-inflammatory factor secretion by promoting the generation of Th2 and Treg cells, along with the activation of M2-type microglia. These approaches may alleviate neuroinflammation and facilitate the repair of neural tissues.
Collapse
Affiliation(s)
- Yuxiao Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zilin Ren
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Juntang Yan
- Library, Beijing University of Chinese Medicine, Beijing, China
| | - Congai Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yanhui He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuyu Shi
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fafeng Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qingguo Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Changxiang Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xueqian Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
46
|
Guo H, Zhao Y, Zhang Z, Xu Y, Chen Y, Lei T, Zhao Y. The Presence and Pathogenic Roles of M(IL-33 + IL-2) Macrophages in Allergic Airway Inflammation. Allergy 2025; 80:1298-1308. [PMID: 39670958 DOI: 10.1111/all.16430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 10/02/2024] [Accepted: 11/06/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Macrophages, one of the most abundant immune cells in the lung, have drawn great attention in allergic asthma. Currently, most studies emphasize alternative activated (M2) polarization bias. However, macrophage function in allergic asthma is still controversial. Interleukin (IL)-9 contributes to the development and pathogenesis of allergic airway inflammation. We sought to investigate the IL-9-producing macrophage and its role in allergic asthma. METHODS The model of ovalbumin (OVA)-induced allergic airway inflammation was employed to evaluate IL-9 production in macrophages of lung tissues. We used 22 cytokines or stimuli to screen for IL-9-producing mouse macrophage subset in vitro. Real-time PCR, flow cytometry, ELISA, and RNA-seq to explore the subset. Conditional IL-33 receptor knockout (Lyz-ST2KO) mice and cellular adoptive transfer experiment were used to characterize the potential roles of M(IL-33 + IL-2) in allergic asthma. RESULTS We identified a unique pathogenic IL-9-producing macrophage in OVA-induced allergic airway inflammation. We found that only IL-33 significantly induced IL-9 production in mouse macrophages, and IL-2 collaborated with IL-33 to promote IL-9 production, referred to as M(IL-33 + IL-2). Importantly, human monocyte-derived macrophages produced IL-9 after IL-33 and IL-2 stimulation. Using Lyz-ST2KO mice and adoptive transfer of M(IL-33 + IL-2), we found that M(IL-33 + IL-2) significantly promoted pathogenesis in OVA-induced allergic airway inflammation. M(IL-33 + IL-2) has a distinctive gene expression profile with high expression of IL-9, IL-5, and IL-13 and its polarization is dependent on JAK2-STAT3-IRF1 pathway. CONCLUSIONS The identification of M(IL-33 + IL-2) subset extends the diversity and heterogeneity of macrophage subsets and may offer novel therapeutic strategies for the treatment of allergic inflammation.
Collapse
Affiliation(s)
- Han Guo
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yang Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Zhaoqi Zhang
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen, China
| | - Yanan Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yifang Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Tong Lei
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
47
|
Fei X, Li N, Xu X, Zhu Y. Macrophage biology in the pathogenesis of Helicobacter pylori infection. Crit Rev Microbiol 2025; 51:399-416. [PMID: 39086061 DOI: 10.1080/1040841x.2024.2366944] [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: 08/03/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 08/02/2024]
Abstract
Infection with H. pylori induces chronic gastric inflammation, progressing to peptic ulcer and stomach adenocarcinoma. Macrophages function as innate immune cells and play a vital role in host immune defense against bacterial infection. However, the distinctive mechanism by which H. pylori evades phagocytosis allows it to colonize the stomach and further aggravate gastric preneoplastic pathology. H. pylori exacerbates gastric inflammation by promoting oxidative stress, resisting macrophage phagocytosis, and inducing M1 macrophage polarization. M2 macrophages facilitate the proliferation, invasion, and migration of gastric cancer cells. Various molecular mechanisms governing macrophage function in the pathogenesis of H. pylori infection have been identified. In this review, we summarize recent findings of macrophage interactions with H. pylori infection, with an emphasis on the regulatory mechanisms that determine the clinical outcome of bacterial infection.
Collapse
Affiliation(s)
- Xiao Fei
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Nianshuang Li
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xinbo Xu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yin Zhu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
48
|
Gauthier T, Lim YJ, Jin W, Liu N, Patiño LC, Chen W, Warren J, Martin D, Morell RJ, Dveksler G, Su GH, Chen W. Activin A activation of Smad3 mitigates innate inflammation in mouse models of psoriasis and sepsis. J Clin Invest 2025; 135:e187063. [PMID: 40067393 PMCID: PMC12043092 DOI: 10.1172/jci187063] [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: 09/13/2024] [Accepted: 03/05/2025] [Indexed: 05/02/2025] Open
Abstract
Phosphorylation of Smad3 is a critical mediator of TGF-β signaling, which plays an important role in regulating innate immune responses. However, whether Smad3 activation can be regulated in innate immune cells in TGF-β-independent contexts remains poorly understood. Here, we show that Smad3 is activated through the phosphorylation of its C-terminal residues (pSmad3C) in murine and human macrophages in response to bacterial and viral ligands, and this activation is mediated by activin A in a TGF-β-independent manner. Specifically, infectious ligands, such as LPS, induced secretion of activin A through the transcription factor STAT5 in macrophages, and activin A signaling in turn activated pSmad3C. This activin A/Smad3 axis controlled mitochondrial ATP production and ATP conversion into adenosine by CD73 in macrophages, enforcing an antiinflammatory mechanism. Consequently, mice with a deletion of activin A receptor 1b specifically in macrophages (Acvr1bfl/fl-Lyz2cre) succumbed more to sepsis as a result of uncontrolled inflammation and exhibited exacerbated skin disease in a mouse model of imiquimod-induced psoriasis. Thus, we have revealed a previously unrecognized natural brake to inflammation in macrophages that occurs through the activation of Smad3 in an activin A-dependent manner.
Collapse
Affiliation(s)
- Thierry Gauthier
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Yun-Ji Lim
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Wenwen Jin
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Na Liu
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Liliana C. Patiño
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Weiwei Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - James Warren
- Department of Pathology, Uniformed Services University, Bethesda, Maryland, USA
| | - Daniel Martin
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Robert J. Morell
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Gabriela Dveksler
- Department of Pathology, Uniformed Services University, Bethesda, Maryland, USA
| | - Gloria H. Su
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
49
|
Ukaegbu K, Allen E, Svoboda KKH. Reactive Oxygen Species and Antioxidants in Wound Healing: Mechanisms and Therapeutic Potential. Int Wound J 2025; 22:e70330. [PMID: 40288766 PMCID: PMC12034374 DOI: 10.1111/iwj.70330] [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: 08/21/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 04/29/2025] Open
Abstract
Wound healing is a complex biological process encompassing haemostasis, inflammation, proliferation and matrix remodelling. Reactive oxygen species (ROS) play a pivotal role in regulating key events such as antimicrobial defence, platelet activation and angiogenesis. However, excessive ROS levels can induce oxidative stress (OS), disrupting the healing cascade and contributing to chronic wounds, inflammation and impaired tissue repair. Systemic conditions like diabetes, obesity, smoking and ageing further exacerbate OS, highlighting its clinical significance in wound management. Antioxidants (AOx), both endogenous and exogenous, have demonstrated therapeutic potential in mitigating OS, promoting wound closure and enhancing cellular recovery. Compounds like Vitamin E, curcumin, ferulic acid and resveratrol improve AOx enzyme activity, reduce oxidative damage and accelerate wound healing in multiple studies. Emerging evidence supports targeting oxidative pathways as a viable strategy to improve outcomes in chronic and systemic OS-related conditions. This review explores the dual role of ROS in wound healing, the impact of OS in systemic diseases, and the therapeutic potential of AOx in fostering optimal healing outcomes, advocating for robust clinical trials to establish standardised interventions.
Collapse
Affiliation(s)
- Kelechi Ukaegbu
- Department of PeriodontologyTexas A&M School of DentistryDallasTexasUSA
- Private PracticeHoustonTexasUSA
| | - Edward Allen
- Department of PeriodontologyTexas A&M School of DentistryDallasTexasUSA
- Center for Advanced Dental EducationDallasTexasUSA
| | - Kathy K. H. Svoboda
- Department of Biomedical SciencesTexas A&M School of DentistryDallasTexasUSA
| |
Collapse
|
50
|
He J, Dai Y, Xu F, Huang X, Gao Y, Liu L, Zhang W, Liu J. High-density lipoprotein-based nanoplatforms for macrophage-targeted diagnosis and therapy of atherosclerosis. Int J Biol Macromol 2025; 306:140826. [PMID: 40010459 DOI: 10.1016/j.ijbiomac.2025.140826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/19/2025] [Accepted: 02/07/2025] [Indexed: 02/28/2025]
Abstract
Atherosclerosis, the primary cause of cardiovascular disease, which has the highest mortality worldwide, is a chronic inflammatory disease mainly induced by excessive lipid accumulation in plaque macrophages. Lipid-laden macrophages are crucial at all stages of atherosclerotic lesion progression and are, thus, regarded as popular therapeutic targets for atherosclerosis. High-density lipoprotein (HDL), an endogenous particle with excellent atherosclerotic plaque-homing properties, is considered a potential therapeutic agent for treating atherosclerosis. Based on the excellent properties of HDL, reconstituted HDL (rHDL), with physiological functions similar to those of its natural counterparts, have been successfully prepared as therapeutics and are also recognized as a potential nanoplatform for delivering drugs or contrast agents to atherosclerotic plaques owing to their high biocompatibility, amphiphilic structure, and macrophage-targeting capability. In this review, we focus on the (a) important role of macrophages in atherosclerotic lesions, (b) biological properties of rHDL as a delivery nanoplatform in atherosclerotic diseases, and (c) multiple applications of rHDL in the diagnosis and treatment of atherosclerosis. We systematically summarize the novel applications of rHDL with unique advantages in atherosclerosis, aiming to provide specific insights and inspire additional innovative research in this field.
Collapse
Affiliation(s)
- Jianhua He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China; School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yingxuan Dai
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Fengfei Xu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Xinya Huang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yu Gao
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Lisha Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|