1
|
Dutta SD, Hexiu J, Moniruzzaman M, Patil TV, Acharya R, Kim JS, Lim KT. Tailoring osteoimmunity and hemostasis using 3D-Printed nano-photocatalytic bactericidal scaffold for augmented bone regeneration. Biomaterials 2025; 316:122991. [PMID: 39662273 DOI: 10.1016/j.biomaterials.2024.122991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
Bone hemorrhage, infection, and large bone defects following surgical treatment of traumatic bone injury have raised potential concerns, underscoring the urgent need to develop multifunctional therapeutic platforms that can effectively address traumatic bone regeneration. Advancements in three-dimensional (3D) printing technology have propelled the development of several engineering disciplines, such as tissue engineering. Nevertheless, 3D-printed frameworks with conventional materials often lack multifunctional capabilities to promote specific activities for diverse regeneration purposes. In this study, we developed a highly oxidized two-dimensional (2D) graphitic carbon nitride (Ox-gCN) as a nano-photocatalyst to reinforce alginate/gelatin (ALG)-based hydrogel scaffolds (ALG/CN) to achieve an anti-inflammatory and osteo-immunomodulatory niche with superior hemostatic ability for traumatic bone injury repair. Sulfuric acid oxidation enhances the oxygen-containing functional groups of the g-CN surface and promotes cell adhesion and differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs) in vitro. Moreover, the excellent visible light-activated photocatalytic characteristics of the ALG/CN scaffold were used in antibacterial studies. In addition, the ALG/CN bio/nanocomposite scaffold facilitates M2 polarization of macrophages than did pristine ALG scaffolds. Furthermore, ALG/CN scaffold induced hBMSCs differentiation by upregulating ERK and MAPKs phosphorylation during osteo-immunomodulation. In a rat calvaria defect model, the fabricated ALG/CN scaffolds induced new bone formation through collagen deposition and activation of osteocalcin proteins without inflammation in vivo. These results highlight the potential of 3D-printed functionalized 2D carbon nitrides in regulating the bone immune microenvironment, which may be beneficial for developing advanced tissue constructs, especially for traumatic bone regeneration in clinical settings.
Collapse
Affiliation(s)
- Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea; Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea; Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Jin Hexiu
- Department of Oral and Maxillofacial Surgery, Capital Medical University, Beijing-1000054, China
| | - Md Moniruzzaman
- Department of Chemical and Biological Engineering, Gachon University, Seongnam, 13120, Republic of Korea
| | - Tejal V Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea; Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Rumi Acharya
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea; Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jong Sung Kim
- Department of Chemical and Biological Engineering, Gachon University, Seongnam, 13120, Republic of Korea.
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea; Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea; Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
2
|
Shi X, Askari Rizvi SF, Yang Y, Liu G. Emerging nanomedicines for macrophage-mediated cancer therapy. Biomaterials 2025; 316:123028. [PMID: 39693782 DOI: 10.1016/j.biomaterials.2024.123028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Tumor-associated macrophages (TAMs) contribute to tumor progression by promoting angiogenesis, remodeling the tumor extracellular matrix, inducing tumor invasion and metastasis, as well as immune evasion. Due to the high plasticity of TAMs, they can polarize into different phenotypes with distinct functions, which are primarily categorized as the pro-inflammatory, anti-tumor M1 type, and the anti-inflammatory, pro-tumor M2 type. Notably, anti-tumor macrophages not only directly phagocytize tumor cells, but also present tumor-specific antigens and activate adaptive immunity. Therefore, targeted regulation of TAMs to unleash their potential anti-tumor capabilities is crucial for improving the efficacy of cancer immunotherapy. Nanomedicine serves as a promising vehicle and can inherently interact with TAMs, hence, emerging as a new paradigm in cancer immunotherapy. Due to their controllable structures and properties, nanomedicines offer a plethora of advantages over conventional drugs, thus enhancing the balance between efficacy and toxicity. In this review, we provide an overview of the hallmarks of TAMs and discuss nanomedicines for targeting TAMs with a focus on inhibiting recruitment, depleting and reprogramming TAMs, enhancing phagocytosis, engineering macrophages, as well as targeting TAMs for tumor imaging. We also discuss the challenges and clinical potentials of nanomedicines for targeting TAMs, aiming to advance the exploitation of nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Xueying Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Syed Faheem Askari Rizvi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China; Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 54000, Punjab, Pakistan
| | - Yinxian Yang
- School of Pharmaceutical Sciences, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| |
Collapse
|
3
|
Kuan CH, Chang L, Ho CY, Tsai CH, Liu YC, Huang WY, Wang YN, Wang WH, Wang TW. Immunomodulatory hydrogel orchestrates pro-regenerative response of macrophages and angiogenesis for chronic wound healing. Biomaterials 2025; 314:122848. [PMID: 39342917 DOI: 10.1016/j.biomaterials.2024.122848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/22/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
Chronic wound healing often encounters challenges characterized by prolonged inflammation and impaired angiogenesis. While the immune response plays a pivotal role in orchestrating the intricate process of wound healing, excessive inflammation can hinder tissue repair. In this study, a bilayer alginate hydrogel system encapsulating polyelectrolyte complex nanoparticles (PCNs) loaded with anti-inflammatory cytokines and angiogenic growth factors is developed to address the challenges of chronic wound healing. The alginate hydrogel is designed using two distinct crosslinking methods to achieve differential degradation, thereby enabling precise spatial and temporal controlled release of PCNs. Initially, interleukin-10 (IL-10) is released to mitigate inflammation, while unsaturated PCNs bind and remove accumulated pro-inflammatory cytokines at the wound site. Subsequently, angiogenic growth factors, including vascular endothelial growth factor and platelet-derived growth factor, are released to promote vascularization and vessel maturation. Our results demonstrate that the bilayer hydrogel exhibits distinct degradation kinetics between the two layers, facilitating the staged release of multiple signaling molecules. In vitro experiments reveal that IL-10 can activate the Jak1/STAT3 pathway, thereby suppressing pro-inflammatory cytokines and chemokines while down-regulating inflammation-related genes. In vivo studies demonstrate that application of the hydrogel in chronic wounds using diabetic murine model promotes healing by positively modulating multiple integral reparative mechanisms. These include reducing inflammation, promoting macrophage polarization towards a pro-regenerative phenotype, enhancing keratinocyte migration, stimulating angiogenesis, and expediting wound closure. In conclusion, our hydrogel system effectively mitigates inflammatory responses and provides essential physiological cues by inducing a synergistic angiogenic effect, thus offering a promising approach for the treatment of chronic wounds.
Collapse
Affiliation(s)
- Chen-Hsiang Kuan
- Division of Plastic Surgery, Department of Surgery, National Taiwan University Hospital, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taiwan.
| | - Ling Chang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Yu Ho
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan; Department of Bioengineering, Rice University, Houston, USA
| | - Chia-Hsuan Tsai
- Division of Plastic Surgery, Department of Surgery, Chang Gung Memorial Hospital, Keelung Branch, Taiwan
| | - Yu-Chung Liu
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan; Department of Biomedical Engineering, University of Michigan-Ann Arbor, Michigan, USA
| | - Wei-Yuan Huang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Ning Wang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Wei-Hung Wang
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Tzu-Wei Wang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
4
|
Huang SH, Wang CC, Shen PC, Liu ZM, Chen SJ, Tien YC, Lu CC. Suramin enhances proliferation, migration, and tendon gene expression of human supraspinatus tenocytes. J Orthop Res 2025; 43:252-263. [PMID: 39358851 DOI: 10.1002/jor.25990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/23/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
Rotator cuff tendinopathy is a common musculoskeletal disorder with limited pharmacological treatment strategies. This study aimed to investigate tenocytes' functional in vitro response from a ruptured supraspinatus tendon to suramin administration and to elucidate whether suramin can enhance tendon repair and modulate the inflammatory response to injury. Tenocytes were obtained from human supraspinatus tendons (n = 6). We investigated the effect of suramin on LPS-induced inflammatory responses and the underlying molecular mechanisms in THP-1 macrophages. Suramin enhanced the proliferation, cell viability, and migration of tenocytes. It also increased the protein expression of PCNA and Ki-67. Suramin-treated tenocytes exhibited increased expression of COL1A1, COL3A1, TNC, SCX, and VEGF. Suramin significantly reduced LPS-induced iNOS, COX2 synthesis, inflammatory cytokine TNF-α production, and inflammatory signaling by influencing the NF-κB pathways in THP-1 cells. Our results suggest that suramin holds great promise as a therapeutic option for treating rotator cuff tendinopathy.
Collapse
Affiliation(s)
- Shih-Hao Huang
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chih-Chien Wang
- Department of Anesthesiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Po-Chih Shen
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zi-Miao Liu
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shu-Jung Chen
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yin-Chun Tien
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Cheng-Chang Lu
- Department of Orthopaedic Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Orthopaedic Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
5
|
Su D, Li M, Xie Y, Xu Z, Lv G, Jiu Y, Lin J, Chang CJ, Chen H, Cheng F. Gut commensal bacteria Parabacteroides goldsteinii-derived outer membrane vesicles suppress skin inflammation in psoriasis. J Control Release 2025; 377:127-145. [PMID: 39532207 DOI: 10.1016/j.jconrel.2024.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/19/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Despite gut microbiota-derived extracellular vesicles (EVs) serving as pivotal mediators in bacteria-host cell interactions, their potential role in modulating skin inflammation remains poorly understood. Here, we developed strategies for mass production of Parabacteroides goldsteinii-derived outer membrane vesicles (Pg OMVs), commonly known as EVs. We found that orally administered Pg OMVs can reach the colon, traverse the intestinal barrier, and circulate to the inflamed skin of psoriasis-like mice, resulting in reduced epidermal hyperplasia, suppressed infiltration of inflammatory cells in the skin lesions, and effective amelioration of both skin and systemic inflammation. Additionally, subcutaneous injection of thermosensitive PF-127 hydrogel loaded with Pg OMVs exerts similar immunomodulatory effects, allowing sustained release of Pg OMVs into skin cells, effectively suppressing skin inflammation and ameliorating symptoms of psoriasis. This study unveils the importance of gut microbiota-derived OMVs, which can target inflamed skin via both the gut-skin axis and local skin administration, providing a promising alternative to live bacteria therapy for the treatment of skin inflammatory diseases.
Collapse
Affiliation(s)
- Dandan Su
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Manchun Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Yuedong Xie
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Zhanxue Xu
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Guowen Lv
- College of Life Sciences, Northwest A&F University, Yangling 712100, China
| | - Yaming Jiu
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Yuquan Road No. 19(A), Shijingshan District, Beijing 100049, China
| | - Jingxiong Lin
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Chih-Jung Chang
- Medical Research Center and Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen 361028, China; School of Medicine, Huaqiao University, Quanzhou 362021, China.
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
6
|
Sato T, Sugiyama D, Koseki J, Kojima Y, Hattori S, Sone K, Nishinakamura H, Ishikawa T, Ishikawa Y, Kato T, Kiyoi H, Nishikawa H. Sustained inhibition of CSF1R signaling augments antitumor immunity through inhibiting tumor-associated macrophages. JCI Insight 2025; 10:e178146. [PMID: 39782686 DOI: 10.1172/jci.insight.178146] [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: 12/04/2023] [Accepted: 11/13/2024] [Indexed: 01/12/2025] Open
Abstract
Tumor-associated macrophages (TAMs) are one of the key immunosuppressive components in the tumor microenvironment (TME) and contribute to tumor development, progression, and resistance to cancer immunotherapy. Several reagents targeting TAMs have been tested in preclinical and clinical studies, but they have had limited success. Here, we show that a unique reagent, FF-10101, exhibited a sustained inhibitory effect against colony-stimulating factor 1 receptor by forming a covalent bond and reduced immunosuppressive TAMs in the TME, which led to strong antitumor immunity. In preclinical animal models, FF-10101 treatment significantly reduced immunosuppressive TAMs and increased antitumor TAMs in the TME. In addition, tumor antigen-specific CD8+ T cells were increased; consequently, tumor growth was significantly inhibited. Moreover, combination treatment with an anti-programmed cell death 1 (anti-PD-1) antibody and FF-10101 exhibited a far stronger antitumor effect than either treatment alone. In human cancer specimens, FF-10101 treatment reduced programmed cell death 1 ligand 1 (PD-L1) expression on TAMs, as observed in animal models. Thus, FF-10101 acts as an immunomodulatory agent that can reduce immunosuppressive TAMs and augment tumor antigen-specific T cell responses, thereby generating an immunostimulatory TME. We propose that FF-10101 is a potential candidate for successful combination cancer immunotherapy with immune checkpoint inhibitors, such as PD-1/PD-L1 blockade.
Collapse
Affiliation(s)
- Takahiko Sato
- Department of Immunology and
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Jun Koseki
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Yasuhiro Kojima
- Laboratory of Computational Life Science, National Cancer Center, Tokyo, Japan
| | - Satomi Hattori
- Department of Immunology and
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Hitomi Nishinakamura
- Division of Cancer Immunology, Research Institute / Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo, Japan
| | | | - Yuichi Ishikawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyoshi Nishikawa
- Department of Immunology and
- Division of Cancer Immunology, Research Institute / Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo, Japan
- Division of Cancer Immune Multicellular System Regulation, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Kindai University Faculty of Medicine, Osaka-sayama, Japan
| |
Collapse
|
7
|
Jia L, Ma M, Xiong W, Zhu J, Cai Y, Chen Y, Jin J, Gao M. Evaluating the Anti-inflammatory Potential of JN-KI3: The Therapeutic Role of PI3Kγ-Selective Inhibitors in Asthma Treatment. Inflammation 2025:10.1007/s10753-024-02180-6. [PMID: 39776396 DOI: 10.1007/s10753-024-02180-6] [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: 03/06/2024] [Revised: 07/16/2024] [Accepted: 10/29/2024] [Indexed: 01/11/2025]
Abstract
Asthma is a chronic airway inflammatory disease of the airways characterized by the involvement of numerous inflammatory cells and factors. Therefore, targeting airway inflammation is one of the crucial strategies for developing novel drugs in the treatment of asthma. Phosphoinositide 3-kinase gamma (PI3Kγ) has been demonstrated to have a significant impact on inflammation and immune responses, thus emerging as a promising therapeutic target for airway inflammatory disease, including asthma. There are few studies reporting on the therapeutic effects of PI3Kγ-selective inhibitors in asthma disease. In this study, we investigated the anti-inflammatory and therapeutic effects of PI3Kγ-selective inhibitor JN-KI3 for treating asthma by utilizing both in vivo and in vitro approaches, thereby proving that PI3Kγ-selective inhibitors could be valuable in the treatment of asthma. In RAW264.7 macrophages, JN-KI3 effectively suppressed C5a-induced Akt phosphorylation in a concentration-dependent manner, with no discernible toxicity observed in RAW264.7 cells. Furthermore, JN-KI3 can inhibit the PI3K/Akt signaling pathway in lipopolysaccharide-induced RAW264.7 cells, leading to the suppression of transcription and expression of the classical inflammatory cytokines in a concentration-dependent manner. Finally, an ovalbumin-induced murine asthma model was constructed to evaluate the initial therapeutic effect of JN-KI3 for treating asthma. Oral administration of JN-KI3 inhibited the infiltration of inflammatory cells and the expression of T-helper type 2 cytokines in bronchoalveolar lavage fluid, which was associated with the suppression of the PI3K signaling pathway. Lung tissue and immunohistochemical studies demonstrated that JN-KI3 inhibited the accumulation of inflammatory cells around the bronchus and blood vessels, as well as the secretion of mucus and excessive deposition of collagen around the airway. In addition, it reduced the infiltration of white blood cells into the lungs. In summary, JN-KI3 shows promise as a candidate for the treatment of asthma. Our study also suggests that the inhibitory effects of PI3Kγ on inflammation could offer an additional therapeutic strategy for pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Lei Jia
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Mengyun Ma
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Wendian Xiong
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Jingyu Zhu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| | - Yanfei Cai
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Jian Jin
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| | - Mingzhu Gao
- Department of Clinical Research Center for Wuxi No.2 People's Hospital, Jiangnan University Medical Center, Wuxi, 214000, Jiangsu, China.
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
8
|
Ohishi K, Dora D, Han CY, Guyer RA, Ohkura T, Kazimierczyk S, Picard N, Leavitt AR, Ott LC, Rahman AA, Mueller JL, Shpigel NY, Jain N, Nagy N, Hotta R, Goldstein AM, Stavely R. Resolving Resident Colonic Muscularis Macrophage Diversity and Plasticity During Colitis. Inflamm Bowel Dis 2025; 31:151-168. [PMID: 39102823 DOI: 10.1093/ibd/izae155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Indexed: 08/07/2024]
Abstract
BACKGROUND Immune cell populations in the intestinal muscularis propria during colitis are poorly resolved. Maintaining homeostasis in this niche is critical, highlighted by the poorer prognosis of inflammatory bowel disease associated with muscularis propria inflammation. METHODS This study utilizes single-cell RNA sequencing to survey the immune cell populations within the muscularis propria of normal colon and dextran sodium sulfate-induced colitis. Findings are validated by immunohistochemistry, flow cytometry and cell-lineage tracing in vivo, and in vitro assays with muscularis macrophages (MMφ). RESULTS In naïve conditions, transcriptional duality is observed in MMφs with 2 major subpopulations: conventional resident Cx3cr1+ MMφs and Lyve1+ MMφs. The Lyve1+ population is phagocytic and expresses several known MMφ markers in mouse and human, confirming their identity as a bona fide MMφ subset. Single-cell transcriptomics indicate that resident MMφs are retained during colitis and exhibit plasticity toward an inflammatory profile. Lyve1+ MMφs, which express anti-inflammatory marker CD163, are absent during colitis, as confirmed by flow cytometry. In contrast, lineage tracing finds that resident Cx3cr1+ MMφs remain during colitis and are not completely replaced by the inflammatory infiltrating monocytes. In vitro studies provide biological evidence of the plasticity of resident Cx3cr1+ MMφs in response to lipopolysaccharide (LPS), mirroring transcriptional observations in vivo of their inflammatory plasticity. Potential markers for colitic MMφs, validated in animal models and in individuals with ulcerative colitis, are identified. CONCLUSIONS Our findings contribute to the understanding of the immune system in the muscularis propria niche during colitis by resolving the heterogeneity and origins of colitic MMφs.
Collapse
Affiliation(s)
- Kensuke Ohishi
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co., Ltd., Akitakata, Hiroshima, Japan
| | - David Dora
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Christopher Y Han
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard A Guyer
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Takahiro Ohkura
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon Kazimierczyk
- Mucosal Immunology and Biology Research Center, Mass General Hospital for Children, Charlestown, MA, USA
| | - Nicole Picard
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Abigail R Leavitt
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Leah C Ott
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ahmed A Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica L Mueller
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nahum Y Shpigel
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Nitya Jain
- Mucosal Immunology and Biology Research Center, Mass General Hospital for Children, Charlestown, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Nandor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Yang P, Rong X, Gao Z, Wang J, Liu Z. Metabolic and epigenetic regulation of macrophage polarization in atherosclerosis: Molecular mechanisms and targeted therapies. Pharmacol Res 2025; 212:107588. [PMID: 39778637 DOI: 10.1016/j.phrs.2025.107588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/05/2024] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
Atherosclerosis, a multifactorial progressive inflammatory disease, is the common pathology underlying cardiovascular and cerebrovascular diseases. The macrophage plasticity is involved in the pathogenesis of atherosclerosis. With the advance of metabolomics and epigenetics, metabolites/metabolic and epigenetic modification such as DNA methylation, histone modification and noncoding RNA, play a crucial role in macrophage polarization and the progression of atherosclerosis. Herein, we provide a comprehensive review of the essential role of metabolic and epigenetic regulation, as well as the crosstalk between the two in regulating macrophage polarization in atherosclerosis. We also highlight the potential therapeutic strategies of regulating macrophage polarization via epigenetic and metabolic modifications for atherosclerosis, and offer recommendations to advance our knowledge of the roles of metabolic-epigenetic crosstalk in macrophage polarization in the context of atherosclerosis. Fundamental studies that elucidate the mechanisms by which metabolic and epigenetic regulation of macrophage polarization influence atherosclerosis will pave the way for novel therapeutic approaches.
Collapse
Affiliation(s)
- Pinglian Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Xiaoling Rong
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhechang Gao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jiaojiao Wang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
10
|
Li C, Li J, Bai Y, Zhang K, Wang Z, Zhang Y, Guan Q, Wang S, Li Z, Li Z, Chen L. Polysialic acid-based nanoparticles for enhanced targeting and controlled dexamethasone release in pulmonary inflammation treatment. Int J Biol Macromol 2025:139550. [PMID: 39778853 DOI: 10.1016/j.ijbiomac.2025.139550] [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: 08/21/2024] [Revised: 12/24/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening conditions characterized by severe inflammation and respiratory failure. Despite the use of dexamethasone (Dex) in treatment, challenges such as poor solubility and systemic side effects persist, highlighting the need for novel therapeutic approaches. This study introduces an innovative nanoparticle delivery system based on chitosan (CS) and polysialic acid (PSA), engineered via electrostatic assembly, to improve the targeted delivery of Dex to inflamed lung tissues. To enhance drug encapsulation and stability, novel taurine-Vitamin E succinate amphiphilic molecules (TVES and TGVES) were synthesized. The unique ability of PSA to specifically target Siglec-1 receptors on M1 macrophages-key contributors to ALI/ARDS-related inflammation-positions this system as a promising strategy for targeted pulmonary therapies. In vitro targeting of M1 macrophages and in vivo reduction of inflammation demonstrate the potential to transform treatment by delivering therapeutic agents precisely to the site of need. This cutting-edge nanoparticle platform not only holds promise for improving ALI/ARDS outcomes but also paves the way for the application of functional additives like taurine in advanced medical therapies.
Collapse
Affiliation(s)
- Cong Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China; Liaoning Key Laboratory for New Drug Development, Shenyang 110036, China
| | - Jing Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Yujie Bai
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Kexin Zhang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Zeyu Wang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Yifan Zhang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Qingyu Guan
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Shiqi Wang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Zehao Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Zhihang Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Lijiang Chen
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China; Liaoning Key Laboratory for New Drug Development, Shenyang 110036, China.
| |
Collapse
|
11
|
Quadros-Pereira L, Nery-Neto JADO, Da Silva EM, Doretto-Silva L, Yariwake VY, Câmara NO, Andrade-Oliveira V. Treatment with sitagliptin exacerbates the M2 phenotype in macrophages in vitro. Int Immunopharmacol 2025; 145:113730. [PMID: 39662268 DOI: 10.1016/j.intimp.2024.113730] [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: 08/19/2024] [Revised: 11/09/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
Macrophages (MØ) participate in the induction and the control of the host's immune response in homeostasis and during inflammatory diseases. Sitagliptin is a drug that inhibits the enzyme dipeptidyl peptidase 4 (DPP-4) and, therefore, increases the bioavailability of the incretins GIP (Gastric inhibitory polypeptide) and GLP-1 (Glucagon-like polypeptide). Thus, sitagliptin has been used to treat obesity and type II diabetes and has recently been associated with anti-inflammatory effects. It is known that the drug can modulate the immune response, however, the underlying mechanisms are not yet completely elucidated, including how they interfere with the activation and function of MØ. Here, we aimed to investigate and characterize the effects of in vitro treatment with sitagliptin on MØ polarization. Bone marrow-derived MØ were differentiated with conditioned medium from the L929 cell line. For M1, MØ were stimulated with IFN-γ and LPS, and for M2, with IL-4 and IL-13 for 24 h. Sitagliptin treatment was performed during MØ polarization. Polarized MØ were assessed for M1/M2 markers, DPP-4, GLP-1 and GIP receptors, mitochondrial dynamics and phagocytosis. Sitagliptin treatment exacerbates the M2 phenotype, featured by increased expression of CD206 and ARG1 and decreased gene expression levels of TNF-α. Sitagliptin-treated M2 altered mitochondrial dynamics with reduced membrane potential and mitochondrial reactive oxygen species production. These differences were accompanied by low gene expression levels of genes related to mitofusion, suggesting that sitagliptin treatment interferes with mitochondria function in M2, and exhibited less phagocytic capacity. In summary, our data suggest that sitagliptin exacerbates M2 profile in vitro.
Collapse
Affiliation(s)
- Laura Quadros-Pereira
- Mucosal Health and Immunology Laboratory (MHIL), Center for Natural and Human Science, Federal University of ABC, Santo André, São Paulo, Brazil
| | - José Arimatéa de Oliveira Nery-Neto
- Mucosal Health and Immunology Laboratory (MHIL), Center for Natural and Human Science, Federal University of ABC, Santo André, São Paulo, Brazil; Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Eloisa Martins Da Silva
- Mucosal Health and Immunology Laboratory (MHIL), Center for Natural and Human Science, Federal University of ABC, Santo André, São Paulo, Brazil; Paulista School of Medicine, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Lorena Doretto-Silva
- Mucosal Health and Immunology Laboratory (MHIL), Center for Natural and Human Science, Federal University of ABC, Santo André, São Paulo, Brazil
| | - Victor Yuji Yariwake
- Mucosal Health and Immunology Laboratory (MHIL), Center for Natural and Human Science, Federal University of ABC, Santo André, São Paulo, Brazil; Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Niels Olsen Câmara
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Vinicius Andrade-Oliveira
- Mucosal Health and Immunology Laboratory (MHIL), Center for Natural and Human Science, Federal University of ABC, Santo André, São Paulo, Brazil; Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil.
| |
Collapse
|
12
|
Ferkel SAM, Holman EA, Sojwal RS, Rubin SJS, Rogalla S. Tumor-Infiltrating Immune Cells in Colorectal Cancer. Neoplasia 2025; 59:101091. [PMID: 39642846 DOI: 10.1016/j.neo.2024.101091] [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: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 12/09/2024]
Abstract
Colorectal cancer encompasses a heterogeneous group of malignancies that differ in pathophysiological mechanisms, immune response and infiltration, therapeutic response, and clinical prognosis. Numerous studies have highlighted the clinical relevance of tumor-infiltrating immune cells among different types of colorectal tumors yet vary in cell type definitions and cell identification strategies. The distinction of immune signatures is particularly challenging when several immune subtypes are involved but crucial to identify novel intercellular mechanisms within the tumor microenvironment. In this review, we compile human and non-human studies on tumor-infiltrating immune cells and provide an overview of immune subtypes, their pathophysiological functions, and their prognostic role in colorectal cancer. We discuss how differentiating immune signatures can guide the development of immunotherapeutic targets and personalized treatment regimens. We analyzed comprehensive human protein biomarker profiles across the entire immune spectrum to improve interpretability and application of tumor studies and to ultimately enhance immunotherapy and advance precision medicine for colorectal cancer patients.
Collapse
Affiliation(s)
- Sonia A M Ferkel
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Elizabeth A Holman
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Raoul S Sojwal
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Samuel J S Rubin
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Stephan Rogalla
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA.
| |
Collapse
|
13
|
Penas FN, Bott E, Carneiro AB, López SA, Torres Bozza P, Goren NB, Gimenez G, Belaunzarán ML. Modified lipids from Trypanosoma cruzi amastigotes down-regulate the pro-inflammatory response and increase the expression of alternative activation markers in macrophages. Microb Pathog 2025; 198:107140. [PMID: 39581235 DOI: 10.1016/j.micpath.2024.107140] [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: 12/12/2023] [Revised: 08/06/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024]
Abstract
Herein, we analyzed the in vitro effect induced by total lipid extracts from Trypanosoma cruzi amastigotes of RA and K98 strains, which were obtained after overnight incubation (RAinc and K98inc) to mimic phospholipid hydrolytic processes that occurred adjacent to degenerating amastigote nests in tissues of Chagas disease patients. We demonstrated that RAinc and K98inc might possess bioactive lipid molecules with anti-inflammatory bias since they inactivated the NF-κB pathway, in contrast to intact lipids. Moreover, different M1/M2 macrophage phenotype markers of polarization were analyzed by RT-qPCR which evidenced that RAinc and K98inc promoted an increased expression of the M2 markers Arginase-1, IL-10, FIZZ and YM-1, and a decreased expression of iNOS and proinflammatory cytokines IL-6 and TNF-α. All these results indicate the relevant role of T. cruzi in bioactive lipid molecules, deepening thus our understanding of their contribution to immunomodulatory mechanisms as well as to macrophage polarization that occurs during the course of Chagas disease.
Collapse
Affiliation(s)
- Federico Nicolas Penas
- Facultad de Medicina, Universidad de Buenos Aires, Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Emanuel Bott
- Facultad de Medicina, Universidad de Buenos Aires, Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM), Buenos Aires, Argentina
| | - Alan Brito Carneiro
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil; Bartholomew Laboratory, Department of Surgery, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Sebastián Andrés López
- Facultad de Medicina, Universidad de Buenos Aires, Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM), Buenos Aires, Argentina
| | - Patricia Torres Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Nora Beatriz Goren
- Facultad de Medicina, Universidad de Buenos Aires, Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Guadalupe Gimenez
- Facultad de Medicina, Universidad de Buenos Aires, Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM), Buenos Aires, Argentina
| | - María Laura Belaunzarán
- Facultad de Medicina, Universidad de Buenos Aires, Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM), Buenos Aires, Argentina.
| |
Collapse
|
14
|
Song J, Wu Y, Chen Y, Sun X, Zhang Z. Epigenetic regulatory mechanism of macrophage polarization in diabetic wound healing (Review). Mol Med Rep 2025; 31:2. [PMID: 39422035 PMCID: PMC11551531 DOI: 10.3892/mmr.2024.13367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Diabetic wounds represent a significant complication of diabetes and present a substantial challenge to global public health. Macrophages are crucial effector cells that play a pivotal role in the pathogenesis of diabetic wounds, through their polarization into distinct functional phenotypes. The field of epigenetics has emerged as a rapidly advancing research area, as this phenomenon has the potential to markedly affect gene expression, cellular differentiation, tissue development and susceptibility to disease. Understanding epigenetic mechanisms is crucial to further exploring disease pathogenesis. A growing body of scientific evidence has highlighted the pivotal role of epigenetics in the regulation of macrophage phenotypes. Various epigenetic mechanisms, such as DNA methylation, histone modification and non‑coding RNAs, are involved in the modulation of macrophage phenotype differentiation in response to the various environmental stimuli present in diabetic wounds. The present review provided an overview of the various changes that take place in macrophage phenotypes and functions within diabetic wounds and discussed the emerging role of epigenetic modifications in terms of regulating macrophage plasticity in diabetic wounds. It is hoped that this synthesis of information will facilitate the elucidation of diabetic wound pathogenesis and the identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Jielin Song
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300000, P.R. China
| | - Yuqing Wu
- The First Clinical Medical College, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Yunli Chen
- The First Clinical Medical College, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Xu Sun
- Department of Traditional Chinese Medicine Surgery, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, P.R. China
| | - Zhaohui Zhang
- Department of Traditional Chinese Medicine Surgery, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, P.R. China
| |
Collapse
|
15
|
Clevenger AJ, Jha A, Moore E, Raghavan SA. Manipulating immune activity of macrophages: a materials and mechanics perspective. Trends Biotechnol 2025; 43:131-144. [PMID: 39155172 PMCID: PMC11717646 DOI: 10.1016/j.tibtech.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024]
Abstract
Macrophage immune cells exist on a plastic spectrum of phenotypes governed by their physical and biochemical environment. Controlling macrophage function to facilitate immunological regeneration or fighting pathology has emerged as a therapeutic possibility. The rate-limiting step in translating macrophage immunomodulation therapies has been the absence of fundamental knowledge of how physics and biochemistry in the macrophage microenvironment converge to inform phenotype. In this review we explore recent trends in bioengineered model systems that integrate physical and biochemical variables applied to macrophage mechanosensing and plasticity. We focus on how tuning of mechanical forces and biomaterial composition orchestrate macrophage function in physiological and pathological contexts. Ultimately, a broader understanding of stimuli-responsiveness in macrophages leads to informed design for future modulatory therapies.
Collapse
Affiliation(s)
- Abigail J Clevenger
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Aakanksha Jha
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Erika Moore
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
| | - Shreya A Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
16
|
Isali I, McClellan P, Wong TR, Hijaz S, Fletcher DR, Liu G, Bonfield TL, Anderson JM, Hijaz A, Akkus O. Differential effects of macrophage subtype-specific cytokines on fibroblast proliferation and endothelial cell function in co-culture system. J Biomed Mater Res A 2025; 113:e37799. [PMID: 39295242 DOI: 10.1002/jbm.a.37799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/25/2024] [Accepted: 09/06/2024] [Indexed: 09/21/2024]
Abstract
Macrophages are involved in several critical activities associated with tissue repair and regeneration. Current approaches in regenerative medicine are focusing on leveraging the innate immune response to accelerate tissue regeneration and improve long-term healing outcomes. Of particular interest in this regard are the currently known, four main M2 macrophage subtypes: M2interleukin (IL)-4,IL-13, M2IC, M2IL-10, M2non-selective adenosine receptor agonists (NECA) (M2IL-4,IL-13 → M2NECA). In this study, rat bone marrow-derived macrophages (M0) were polarized to each of the four subtypes M2IL-4,IL-13 → M2NECA and cultured for 72 h in vitro. Luminex assay results highlighted increased production of tissue inhibitor of metalloproteinases-1 (TIMP-1) for M2IL-4,IL-13, higher amounts of transforming growth factor-beta 1 (TGF-β1) for M2IL-10, and elevated vascular endothelial growth factor A (VEGF-A) from M2NECA. Co-culture experiments performed with M2IL-10 macrophages and L929 fibroblasts highlighted the increased production of soluble collagen within the media as well as higher amounts of collagen in the extracellular matrix. Human umbilical vein endothelial cells (HUVECs) were co-cultured with M2NECA macrophages, which demonstrated an increase in intercellular adhesion molecule (ICAM) and platelet endothelial cell adhesion molecule (PECAM), as well as increased formation of endothelial tubes. The findings of this study emphasize a critical demand for further characterization and analyses of distinct M2 subtypes and careful selection of specific macrophage populations for regeneration of specific tissue types. The current, broad classification of "M2" may be sufficient in many general tissue engineering applications, but, as conditions are constantly in flux within the microenvironment in vivo, a higher degree of specificity and control over the initial M2 subtype could result in more consistent long-term outcomes where macrophages are utilized as part of an overall regenerative strategy.
Collapse
Affiliation(s)
- Ilaha Isali
- Department of Urology, Weill Cornell Medicine, New York, New York, USA
| | - Phillip McClellan
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Thomas R Wong
- Department of Urology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sara Hijaz
- Department of Urology, Case Western Reserve University, Cleveland, Ohio, USA
| | - David R Fletcher
- Case Western Reserve University, Department of Genetics and Genome Sciences, Cleveland, Ohio, USA
| | - Guiming Liu
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, Ohio, USA
| | - Tracey L Bonfield
- Case Western Reserve University, Department of Genetics and Genome Sciences, Cleveland, Ohio, USA
| | - James M Anderson
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Adonis Hijaz
- Department of Urology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ozan Akkus
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Orthopedics, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
17
|
Illingworth EJ, Rychlik KA, Maertens A, Sillé FCM. Sex-specific transcriptomic effects of low-dose inorganic arsenic exposure on bone marrow-derived macrophages. Toxicology 2025; 510:153988. [PMID: 39515575 DOI: 10.1016/j.tox.2024.153988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/19/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Both tissue-resident macrophages and monocytes recruited from the bone marrow that transform into tissue-resident cells play critical roles in mediating homeostasis as well as in the pathology of inflammatory diseases. Inorganic arsenic (iAs) is the most common drinking water contaminant worldwide and represents a major public health concern. There are numerous diseases caused by iAs exposure in which macrophages are involved, including cardiovascular disease, cancer, and increased risk of (respiratory) infectious diseases. Notably, prenatal iAs exposure is also associated with negative birth outcomes and developmental immunotoxicity (DIT) contributing to long-term adverse outcomes of these immune-related diseases. Therefore, understanding the effects of iAs exposure on macrophages, particularly during immune development or tissue injury and inflammation, can help us better grasp the full range of arsenic immunotoxicity and better design therapeutic targets for iAs-induced diseases particularly in exposed populations. In contrast to prior published studies which often only focused on the effect of iAs on mature macrophages after development, in this study, we analyzed the transcriptome of M0-, M1- and M2-polarized male and female murine bone marrow-derived macrophages (BMDMs) which were exposed to iAs during the differentiation phase, as a model to study iAs (developmental) immunotoxicity. We identified differentially expressed genes by iAs in a sex- and stimulation-dependent manner and used bioinformatics tools to predict protein-protein interactions, transcriptional regulatory networks, and associated biological processes. Overall, our data suggest that M1-stimulated, especially female-derived, BMDMs are most susceptible to iAs exposure during differentiation. Most notably, we observed significant downregulation of major proinflammatory transcription factors, like IRF8, and its downstream targets, as well as genes encoding proteins involved in pattern recognition and antigen presentation, such as TLR7, TLR8, and H2-D1, potentially providing causal insight regarding the role of (early-life) arsenic exposure in perturbing immune responses to infectious diseases. We also observed significant downregulation of genes involved in processes crucial to coordinating a proinflammatory response including leukocyte migration, differentiation, and cytokine and chemokine production and response. Finally, we discovered that 24 X-linked genes were dysregulated in iAs-exposed female stimulation groups compared to only 3 across the iAs-exposed male stimulation groups. These findings elucidate the potential mechanisms underlying the sex-differential iAs-associated immune-related disease risk.
Collapse
Affiliation(s)
- Emily J Illingworth
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Kristal A Rychlik
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Public Health Program, School of Health Professions, Mayborn College of Health Sciences, University of Mary Hardin-Baylor, Belton, TX, USA
| | - Alexandra Maertens
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Fenna C M Sillé
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
18
|
Gudgeon J, Dannoura A, Chatterjee R, Sidgwick F, Raymond BB, Frey AM, Marin-Rubio JL, Trost M. Mass spectrometry-based proteomic exploration of diverse murine macrophage cellular models. Life Sci Alliance 2025; 8:e202402760. [PMID: 39510801 PMCID: PMC11544424 DOI: 10.26508/lsa.202402760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024] Open
Abstract
Immortalised cell lines that mimic their primary cell counterparts are fundamental to research, particularly when large cell numbers are required. Here, we report that immortalisation of bone marrow-derived macrophages (iBMDMs) using the J2 virus resulted in the loss of a protein of interest, MSR1, in WT cells by an unknown mechanism. This led us to perform an in-depth mass spectrometry-based proteomic characterisation of common murine macrophage cell lines (J774A.1, RAW264.7, and BMA3.1A7), in comparison with the iBMDMs, as well as primary BMDMs from both C57BL/6 and BALB/c mice. This analysis revealed striking differences in protein profiles associated with macrophage polarisation, phagocytosis, pathogen recognition, and interferon signalling. Among the cell lines, J774A.1 cells were the most similar to the gold standard primary BMDM model, whereas BMA3.1A7 cells were the least similar because of the reduction in abundance of several key proteins related closely to macrophage function. This comprehensive proteomic dataset offers valuable insights into the use and suitability of macrophage cell lines for cell signalling and inflammation research.
Collapse
Affiliation(s)
- Jack Gudgeon
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Abeer Dannoura
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ritika Chatterjee
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Frances Sidgwick
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Andrew M Frey
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Matthias Trost
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
19
|
Hu D, Yang Z, Zhang J, Liu G, Pi J, Xu J, Wang Y, Zhao Y. Copper homeostasis; A rapier between mycobacteria and macrophages. FASEB Bioadv 2025; 7:e1484. [PMID: 39781425 PMCID: PMC11705462 DOI: 10.1096/fba.2024-00166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Copper is a vital trace element crucial for mediating interactions between Mycobacterium and macrophages. Within these immune cells, copper modulates oxidative stress responses and signaling pathways, enhancing macrophage immune functions and facilitating Mycobacterium clearance. Conversely, copper may promote Mycobacterium escape from macrophages through various mechanisms: inhibiting macrophage activity, diminishing phagocytic and bactericidal capacities, and supporting Mycobacterium survival and proliferation. This paradox has intensified research focus on the regulatory role of copper in immune cell-pathogen interactions. Interactions among metal ions can affect Mycobacterium concentration, distribution, and activity within an organism. In this review, we have elucidated the role of copper in these interactions, focusing on the mechanisms by which this metal influences both the immune defense mechanisms of macrophages and the survival strategies of Mycobacterium. The findings suggest that manipulating copper levels could enhance macrophage bactericidal functions and potentially limit Mycobacterium resistance. Therefore, elucidating the regulatory role of copper is pivotal for advancing our understanding of metal homeostasis in immune cell-pathogen dynamics and TB pathogenesis. Furthermore, we recommend further investigation into the role of copper in TB pathogenesis to advance tuberculosis diagnosis and treatment and gain comprehensive insights into metal homeostasis in infectious disease contexts.
Collapse
Affiliation(s)
- Di Hu
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
- Institute of Laboratory Medicine, School of Medical TechnologyGuangdong Medical UniversityDongguanGuangdongChina
| | - Zisha Yang
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
- Institute of Laboratory Medicine, School of Medical TechnologyGuangdong Medical UniversityDongguanGuangdongChina
| | - Jun‐ai Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
| | - Ganbin Liu
- Department of RespirationDongguan 6th HospitalDongguanGuangdongChina
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
- Institute of Laboratory Medicine, School of Medical TechnologyGuangdong Medical UniversityDongguanGuangdongChina
| | - Junfa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
- Institute of Laboratory Medicine, School of Medical TechnologyGuangdong Medical UniversityDongguanGuangdongChina
| | - Yan Wang
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
- Microbiology and Immunology DepartmentGuangdong Medical UniversityDongguanGuangdongChina
| | - Yi Zhao
- Guangdong Provincial Key Laboratory of Medical Molecular DiagnosticsThe First Dongguan Affiliated Hospital, Guangdong Medical UniversityDongguanGuangdongChina
- Institute of Laboratory Medicine, School of Medical TechnologyGuangdong Medical UniversityDongguanGuangdongChina
- Microbiology and Immunology DepartmentGuangdong Medical UniversityDongguanGuangdongChina
| |
Collapse
|
20
|
Yang K, Lu Y, Gu J, Nie Y, Zhang T. Identifying novel aging-related diagnostic and prognostic models and aging-targeted drugs for sepsis patients. Sci Rep 2024; 14:31445. [PMID: 39732977 DOI: 10.1038/s41598-024-83111-1] [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: 09/22/2024] [Accepted: 12/11/2024] [Indexed: 12/30/2024] Open
Abstract
Sepsis is defined as a dysfunctional, life-threatening response to infection leading to multiorgan dysfunction and failure. During the past decade, studies have highlighted the relationship between sepsis and aging. However, the role of aging-related mechanisms in the progression and prognosis of sepsis remains unclear. In the present study, we divided sepsis patients into High- and Low-aging groups based on the gene set variation analysis (GSVA) scores of GOBP-AGING gene set. Sepsis patients in the high-aging group exhibited higher levels of infiltration of innate immune cells, lower levels of infiltration of adaptive immune cells, and a worse prognosis than those in the Low-aging group. Additionally, the MPO to MME ratio (MPO/MME) appears to be an effective biomarker for predicting the prognosis of sepsis patients. Moreover, ARG1/SEC63 and ARG1/CDKN1C appear to be effective and robust biomarkers for the early diagnosis of sepsis patients. Finally, we found that thalidomide (TAL) significantly ameliorated LPS induced inflammation and organ injury and attenuated LPS induced cellular senescence in lung and kidney. Overall, this study provides new insights into the heterogeneity of sepsis, reveals the vital role of aging-related markers in the prognosis and diagnosis of sepsis and demonstrates that TAL is a novel aging-targeted drug for sepsis patients by attenuating LPS induced cellular senescence.
Collapse
Affiliation(s)
- Kai Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Ministry of Education, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yaoyao Lu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jian Gu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Ministry of Education, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yingli Nie
- Department of Dermatology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China.
| | - Tao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, Ministry of Education, Huazhong University of Science and Technology, Wuhan, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
21
|
Hu S, Kang H, Bae M, Kim MB, Jang H, Corvino O, Pham TX, Lee Y, Smyth JA, Park YK, Lee JY. Histone Deacetylase 9 Deletion Inhibits Hepatic Steatosis and Adipose Tissue Inflammation in Male Diet-Induced Obese Mice. J Gastroenterol Hepatol 2024. [PMID: 39730208 DOI: 10.1111/jgh.16856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 12/29/2024]
Abstract
AIM The goal of this study was to determine the role of histone deacetylase 9 (HDAC9) in the development of diet-induced metabolic dysfunction-associated steatohepatitis (MASH) and white adipose tissue (WAT) dysfunctions. METHODS We fed male and female mice with global Hdac9 knockout (KO) and their wild-type (WT) littermates an obesogenic high-fat/high-sucrose/high-cholesterol (35%/34%/2%, w/w) diet for 20 weeks. RESULTS Hdac9 deletion markedly inhibited body weight gain and liver steatosis with lower liver weight and triglyceride content than WT in male mice but not females. Consistently, hepatic expression of genes crucial for de novo lipogenesis was markedly suppressed only in male, but not female, Hdac9 KO mice. However, Hdac9 deletion had a minimal effect on hepatic inflammation and fibrosis. In WAT, Hdac9 KO showed less adipocyte hypertrophy, inflammation, and fibrosis in male mice compared with WT. In addition, indirect calorimetry demonstrated that male Hdac9 KO mice had significantly higher metabolic rates, respiratory exchange ratios, and energy expenditure without altering physical activities than WT, which was not observed in female mice. CONCLUSIONS Our findings indicate that global deletion of Hdac9 prevented the development of obesity, hepatic steatosis, and WAT inflammation and fibrosis in male mice with diet-induced obesity and MASH, suggesting that a sex-dependent role of HDAC9 may exist in the pathways mentioned above.
Collapse
Affiliation(s)
- Siqi Hu
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Hyunju Kang
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
- Department of Food and Nutrition, Keimyung University, Daegu, South Korea
| | - Minkyung Bae
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
- Department of Food and Nutrition, Yonsei University, Seoul, South Korea
| | - Mi-Bo Kim
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Hyungryun Jang
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Olivia Corvino
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Tho X Pham
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Yoojin Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Joan A Smyth
- Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, Connecticut, USA
| | - Young-Ki Park
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Ji-Young Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
22
|
Li YT, Takaki E, Ouchi Y, Tamai K. Guided monocyte fate to FRβ/CD163 + S1 macrophage antagonises atopic dermatitis via fibroblastic matrices in mouse hypodermis. Cell Mol Life Sci 2024; 82:14. [PMID: 39720957 DOI: 10.1007/s00018-024-05543-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/21/2024] [Accepted: 12/06/2024] [Indexed: 12/26/2024]
Abstract
Macrophages are versatile myeloid leukocytes with flexible cellular states to perform diverse tissue functions beyond immunity. This plasticity is however often hijacked by diseases to promote pathology. Scanning kinetics of macrophage states by single-cell transcriptomics and flow cytometry, we observed atopic dermatitis drastically exhausted a resident subtype S1. Characterized by FRβ/CD163 expression, S1 exhibited strong efferocytosis and chemoattracted monocytes and eosinophils. Here we have delineated mechanisms regulating monocyte decision to acquire S1 identity in skin. During M-CSF driven macrophage differentiation in healthy skin, FRβ was expressed via intrinsic control of STAT6 and ALK5 activities, and did not require heterotypic cellular crosstalk. In contrast, CD163 expression required exposure to fibroblastic secretion. This process depended on SHP1 activity and involved STAT5 inactivation. Suppressed STAT5 activity caused CD163 expression and rendered macrophage insensitive to further induction by fibroblasts. Parsing coculture experiments with in silico ligand expression, we identified laminin-α2 and type-V collagen secreted by hypodermal fibroblasts as CD163-driving factors. S1 identity loss in AD followed a stepwise cascade: reduced laminins availability first dampened CD163 expression, IL4 and TGFβ subsequently acted on CD163lo/- cells to downregulate FRβ. In AD skin, we showed that imitating this fibroblast-macrophage crosstalk with exogenous laminin-211 encouraged monocyte differentiation to S1 macrophages, fostered homeostatic commitment of extravasated eosinophils, and alleviated dermatitis. Hence, we demonstrated that reinforcing a steady-state cue from hypodermal fibroblasts could override maladaptive pressure on macrophage and restored tissue homeostasis.
Collapse
MESH Headings
- Dermatitis, Atopic/metabolism
- Dermatitis, Atopic/pathology
- Receptors, Cell Surface/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Animals
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Macrophages/metabolism
- Monocytes/metabolism
- Mice
- Cell Differentiation
- STAT6 Transcription Factor/metabolism
- Skin/metabolism
- Skin/pathology
- Receptor, Transforming Growth Factor-beta Type I/metabolism
- Receptor, Transforming Growth Factor-beta Type I/genetics
- Mice, Inbred C57BL
- Macrophage Colony-Stimulating Factor/metabolism
- Eosinophils/metabolism
- Laminin/metabolism
Collapse
Affiliation(s)
- Yu-Tung Li
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.
| | | | - Yuya Ouchi
- StemRIM Inc., Ibaraki, Osaka, 567-0085, Japan
| | - Katsuto Tamai
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
23
|
Wu J, Tang J, Zhang L, Wang W, Li Z, Zhou L, Jiang X, Huang Y, Guo Q, Wang W, Ding Z, Cai F, Xi K, Gu Y, Chen L. Biomimetic "Trojan Horse" Fibers Modulate Innate Immunity Cascades for Nerve Regeneration. ACS NANO 2024. [PMID: 39708371 DOI: 10.1021/acsnano.4c12036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2024]
Abstract
Neutrophil membrane vesicles (NMVs) have been successfully applied to control the inflammatory cascade after spinal cord injury (SCI) by acting as an inflammatory factor decoy to front-load the overall inflammation regulatory window; however, the mechanisms by which NMVs regulate macrophage phenotypic shifts as well as their outcomes have rarely been reported. In this study, we demonstrated the "efferocytosis-like" effect of NMVs endocytosed by macrophages, supplementing the TCA cycle intermediate metabolite α-KG by promoting glutamine metabolism, which in turn facilitates oxidative phosphorylation and inhibits the NF-κB signaling pathway to reprogram inflammatory macrophages to the pro-regenerative phenotype. Based on these findings, a "Trojan horse" composite fiber scaffold was constructed; this comprised a carboxylated poly-l-lactic acid shell encapsulated with NMVs and a core loaded with brain-derived neurotrophic factor to spatiotemporally modulate the inflammatory microenvironment by 39.23% and sustainably promote nerve regeneration by 85.67%. In vivo experiments further confirmed the effect of NMV-coated fiber scaffolds on the regulation of early innate immune inflammation and the continuous promotion of nerve regeneration. This study not only further unravels the mechanism of neutrophil membrane-macrophage interactions but also provides a strategy for coordinating inflammatory reprogramming and nerve regeneration following SCI.
Collapse
Affiliation(s)
- Jie Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Jincheng Tang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Lichen Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Wei Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Ziang Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Liang Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Xinzhao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Yiyang Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Qiangqiang Guo
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Wenbo Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Zhouye Ding
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Feng Cai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Yong Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| |
Collapse
|
24
|
Wadhonkar K, Das S, Subramanian R, Sk MH, Singh Y, Baig MS. The effect of cancer cell-derived exosomal proteins on macrophage polarization: An in-depth review. Exp Cell Res 2024; 444:114393. [PMID: 39710293 DOI: 10.1016/j.yexcr.2024.114393] [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: 10/04/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 12/24/2024]
Abstract
Cancer is characterized by unregulated cell proliferation, enabling it to invade and spread to different organs and tissues in the body. Cancer progression is intricately influenced by the complex dynamics within the tumor microenvironment (TME). The TME is a composite and dynamic network comprising cancer cells and various immune cells, including tumor-associated macrophages. Exosomes facilitate the communication between different cancer cells as well as other types of cells. This review particularly focuses on exosomal proteins derived from different cancer cells in mounting the complex crosstalk between cells of cancer and macrophages within the TME. Most cancer-derived exosomal proteins polarize macrophages towards M2 phenotype, promoting cancer aggressiveness, while a few have role switching towards the M1 phenotype, inhibiting cancer proliferation, respectively. In this review, we summarize, for the first time, the dual impact of cancer cell-derived exosomal proteins on macrophage polarization and the associated signaling pathways, offering valuable insights for developing innovative therapeutic strategies against diverse cancer types.
Collapse
Affiliation(s)
- Khandu Wadhonkar
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Soumalya Das
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | | | - Mobbassar Hassan Sk
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK; Institute for Energy and Environmental Flows, University of Cambridge, Cambridge, UK
| | - Yashi Singh
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India.
| |
Collapse
|
25
|
Wang Y, Zhang Y, Ma M, Zhuang X, Lu Y, Miao L, Lu X, Cui Y, Cui W. Mechanisms underlying the involvement of peritoneal macrophages in the pathogenesis and novel therapeutic strategies for dialysis-induced peritoneal fibrosis. Front Immunol 2024; 15:1507265. [PMID: 39749340 PMCID: PMC11693514 DOI: 10.3389/fimmu.2024.1507265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Long-term exposure of the peritoneum to peritoneal dialysate results in pathophysiological changes in the anatomical organization of the peritoneum and progressive development of peritoneal fibrosis. This leads to a decline in peritoneal function and ultrafiltration failure, ultimately necessitating the discontinuation of peritoneal dialysis, severely limiting the potential for long-term maintenance. Additionally, encapsulating peritoneal sclerosis, a serious consequence of peritoneal fibrosis, resulting in patients discontinuing PD and significant mortality. The causes and mechanisms underlying peritoneal fibrosis in patients undergoing peritoneal dialysis remain unknown, with no definitive treatment available. However, abnormal activation of the immune system appears to be involved in altering the structure of the peritoneum and promoting fibrotic changes. Macrophage infiltration and polarization are key contributors to pathological injury within the peritoneum, showing a strong correlation with the epithelial-to-mesenchymal transition of mesothelial cells and driving the process of fibrosis. This article discusses the role and mechanisms underlying macrophage activation-induced peritoneal fibrosis resulting from PD by analyzing relevant literature from the past decade and provides an overview of recent therapeutic approaches targeting macrophages to treat this condition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yingchun Cui
- Department of Nephrology, Second Hospital of Jilin University,
Changchun, China
| | - Wenpeng Cui
- Department of Nephrology, Second Hospital of Jilin University,
Changchun, China
| |
Collapse
|
26
|
Mu H, Yang B, Wang Y, Wang S, Yu W, Jia M, Dong W, Wang X, Xu X, Dong Z, Yang B, Li X, Wang J. Inhibition of fibulin-3 ameliorates periodontal inflammation through reducing M1 macrophage polarization via EGFR/PI3K/AKT pathway. J Periodontol 2024. [PMID: 39692480 DOI: 10.1002/jper.24-0405] [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: 06/25/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND This study aimed to evaluate the role of fibulin-3 (FBLN3) in macrophage polarization, its mechanism, and its effect on periodontitis. METHODS We conducted studies on periodontitis using both clinical samples and ligature-induced mouse periodontitis model. The inflammatory state was assessed using microcomputed tomography, hematoxylin and eosin staining, immunohistochemical staining, and immunofluorescence staining. In vitro, bone marrow-derived macrophages, and RAW 264.7 macrophages were treated with lipopolysaccharide (LPS) and interleukin (IL)-4 to induce polarization. The role of FBLN3 in macrophage polarization was investigated using overexpression plasmids or siRNAs. Furthermore, local injection of adeno-associated virus was employed to suppress FBLN3 expression in periodontal tissues. RESULTS FBLN3 levels were greater in periodontitis tissues. FBLN3 promoted M1 polarization and suppressed M2 polarization in macrophages. The overexpression of FBLN3 promoted M1 polarization via the EGFR/PI3K/AKT signaling pathway, an effect that the epidermal growth factor receptor (EGFR) inhibitor PD153035 reversed. Suppressing FBLN3 expression improved periodontal inflammation and reduced alveolar bone loss in periodontitis. CONCLUSIONS FBLN3 suppression can mitigate periodontitis by decreasing the M1 macrophage ratio. FBLN3 regulates M1 macrophage polarization through the EGFR/PI3K/AKT signaling pathway. PLAIN LANGUAGE SUMMARY Disruption in the collaboration between extracellular matrix (ECM) and immune system is a significant pathology in periodontitis. Macrophages are a crucial part of the immune system and have unique functions, such as polarization. Fibulin-3, an ECM protein, may play a vital role in this dynamic interplay. Fibulin-3 expression is elevated in periodontitis and is closely related to immune cell function. Inhibiting fibulin-3 can alleviate periodontitis by reducing infiltration of immune cells and M1 macrophage ratio. Furthermore, fibulin-3 promoted macrophage M1 polarization by activating the PI3K/AKT signaling pathway through EGFR binding. Our findings offer a clinically relevant rationale for immune response modulation through fibulin-3.
Collapse
Affiliation(s)
- Hailin Mu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Beining Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Yan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Shuo Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Wenqian Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Meie Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Wei Dong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Xinyi Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Xiaoxiao Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Zhipeng Dong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Baochen Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Xuemei Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Jiawei Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
27
|
Kovacs T, Cs. Szabo B, Kothalawala RC, Szekelyhidi V, Nagy P, Varga Z, Panyi G, Zakany F. Inhibition of the H V1 voltage-gated proton channel compromises the viability of human polarized macrophages in a polarization- and ceramide-dependent manner. Front Immunol 2024; 15:1487578. [PMID: 39742270 PMCID: PMC11685079 DOI: 10.3389/fimmu.2024.1487578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
The human voltage-gated proton channel (HV1) provides an efficient proton extrusion pathway from the cytoplasm contributing to the intracellular pH regulation and the oxidative burst. Although its pharmacological inhibition was previously shown to induce cell death in various cell types, no such effects have been examined in polarized macrophages albeit HV1 was suggested to play important roles in these cells. This study highlights that 5-chloro-2-guanidinobenzimidazole (ClGBI), the most widely applied HV1 inhibitor, reduces the viability of human THP-1-derived polarized macrophages at biologically relevant doses with M1 macrophages being the most, and M2 cells the least sensitive to this compound. ClGBI may exert this effect principally by blocking HV1 since the sensitivity of polarized macrophages correlates well with their HV1 expression levels; inhibitors of other macrophage ion channels that may be susceptible for off-target ClGBI effects cause no viability reductions; and Zn2+, another non-specific HV1 blocker, exerts similar effects. As a potential mechanism behind the ClGBI-induced cell death, we identify a complex pH dysregulation involving acidification of the cytoplasm and alkalinization of the lysosomes, which eventually result in membrane ceramide accumulation. Furthermore, ClGBI effects are alleviated by ARC39, a selective acid sphingomyelinase inhibitor supporting the unequivocal significance of ceramide accumulation in the process. Altogether, our results suggest that HV1 inhibition leads to cellular toxicity in polarized macrophages in a polarization-dependent manner, which occurs due to a pH dysregulation and concomitant ceramide overproduction mainly depending on the activity of acid sphingomyelinase. The reduced macrophage viability and plausible concomitant changes in homeostatic M1-M2 balance could contribute to both the therapeutic and potential side effects of HV1 inhibitors that show great promise in the treatment of neuroinflammation and malignant diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Florina Zakany
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
28
|
Chen Y, Zhang X, Huang S, Febbraio M. Hidden features: CD36/SR-B2, a master regulator of macrophage phenotype/function through metabolism. Front Immunol 2024; 15:1468957. [PMID: 39742252 PMCID: PMC11685046 DOI: 10.3389/fimmu.2024.1468957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/30/2024] [Indexed: 01/03/2025] Open
Abstract
Once thought to be in a terminally differentiated state, macrophages are now understood to be highly pliable, attuned and receptive to environmental cues that control and align responses. In development of purpose, the centrality of metabolic pathways has emerged. Thus, macrophage inflammatory or reparative phenotypes are tightly linked to catabolic and anabolic metabolism, with further fine tuning of specific gene expression patterns in specific settings. Single-cell transcriptome analyses have revealed a breadth of macrophage signatures, with some new influencers driving phenotype. CD36/Scavenger Receptor B2 has established roles in immunity and lipid metabolism. Macrophage CD36 is a key functional player in metabolic expression profiles that determine phenotype. Emerging data show that alterations in the microenvironment can recast metabolic pathways and modulate macrophage function, with the potential to be leveraged for therapeutic means. This review covers recent data on phenotypic characterization of homeostatic, atherosclerotic, lipid-, tumor- and metastatic-associated macrophages, with the integral role of CD36 highlighted.
Collapse
Affiliation(s)
- Yuge Chen
- Mike Petryk School of Dentistry, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| | - Xuejia Zhang
- Mike Petryk School of Dentistry, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Maria Febbraio
- Mike Petryk School of Dentistry, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
29
|
Liu Y, Zhao Y, Guo S, Qin D, Yan J, Cheng H, Zhou J, Ren J, Sun L, Peng H, Wu X, Li B. Copper doped carbon dots modified bacterial cellulose with enhanced antibacterial and immune regulatory functions for accelerating wound healing. Carbohydr Polym 2024; 346:122656. [PMID: 39245512 DOI: 10.1016/j.carbpol.2024.122656] [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: 03/18/2024] [Revised: 08/18/2024] [Accepted: 08/23/2024] [Indexed: 09/10/2024]
Abstract
The microenvironment of wound healing is susceptible to bacterial infection, chronic inflammation, oxidative stress, and inadequate angiogenesis, requiring the development of innovative wound dressings with antibacterial, anti-inflammatory, antioxidant, and angiogenic capabilities. This research crafted a new multifunctional bacterial cellulose composite membrane infused with copper-doped carbon dots (BC/Cu(II)-RCDs). Findings validated the successful loading of copper-doped carbon dots onto the BC membrane via hydrogen bonding interactions. Compared to the pure BC membrane, the BC/Cu(II)-RCDs composite membrane exhibited significantly enhanced hydrophilicity, tensile properties, and thermal stability. Diverse in vitro assays demonstrated excellent biocompatibility and antibacterial activity of BC/Cu(II)-RCDs composite membranes, alongside their ability to expedite the inflammatory phase and stimulate angiogenesis. In vivo trials corroborated the membrane's ability to foster epithelial regeneration, collagen deposition, and tissue regrowth in full-thickness skin wounds in rats while also curbing inflammation in infected full-thickness skin wounds. More importantly, the treatment of the BC/Cu(II)-RCDs composite membrane may result in the activation of VEGF and MAPK signaling proteins, which are key players in cell migration, angiogenesis, and skin tissue development. In essence, the developed BC/Cu(II)-RCDs composite membrane shows promise for treating infected wounds and serves as a viable alternative material for medicinal bandages.
Collapse
Affiliation(s)
- Yingyu Liu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, China
| | - Yifan Zhao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, China
| | - Susu Guo
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, China; Academy of Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Danlei Qin
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, China
| | - Jingyu Yan
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, China
| | - Huaiyi Cheng
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, China
| | - Jian Zhou
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Jianing Ren
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, China
| | - Lingxiang Sun
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, China
| | - Hongyi Peng
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, China
| | - Xiuping Wu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, China.
| | - Bing Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, Shanxi, China; Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, Shanxi, China.
| |
Collapse
|
30
|
Zhang N, Zhao L, Li J, Li H, Chen Y. Harnessing Nanotechnology for Gout Therapy: Colchicine-Loaded Nanoparticles Regulate Macrophage Polarization and Reduce Inflammation. Biomater Res 2024; 28:0089. [PMID: 39665079 PMCID: PMC11632155 DOI: 10.34133/bmr.0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/13/2024] [Accepted: 09/24/2024] [Indexed: 12/13/2024] Open
Abstract
Gout is a disease caused by hyperuricemia, characterized by inflammation reactions triggered by macrophage polarization. Colchicine is a commonly used drug for gout treatment, but its mechanism of action remains unclear. The aim of this study was to investigate the regulatory effect of colchicine on macrophage polarization to enhance the therapeutic effectiveness against gout inflammation. To accomplish this, a mouse model was established, and peripheral blood mononuclear cell samples were collected. Single-cell RNA sequencing was employed to reveal cellular heterogeneity and identify key genes. Molecular docking and experimental validation were performed to confirm the binding between the key genes and colchicine. Lentiviral intervention and biochemical indicator detection were conducted to assess the impact of key genes on gout mice. Additionally, the therapeutic effect of colchicine incorporated into neutrophil membrane-coated nanoparticles was investigated. The study found that macrophage polarization plays a critical role in gout, and AHNAK was identified as the key gene through which colchicine affects macrophage polarization. Lentiviral intervention to decrease AHNAK expression was shown to alleviate joint swelling in gout mice and regulate macrophage polarization. Colchicine encapsulated in R4F peptide-modified neutrophil membrane-coated Pluronic F127 nanoparticle (R4F-NM@F127) nanocarriers inhibited M1 macrophage polarization, induced M2 macrophage polarization, alleviated gout, and minimized toxicity to normal tissues. Colchicine suppressed M1 macrophage polarization and induced M2 macrophage polarization by binding to AHNAK protein, thereby alleviating gout. Colchicine incorporated into R4F-NM@F127 nanocarriers can serve as a targeted therapeutic drug to regulate macrophage polarization, alleviate gout, and reduce toxicity to normal tissues.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Rheumatology and Immunology,
Shengjing Hospital Affiliated to China Medical University, Shenyang 110000, China
| | - Lanqing Zhao
- Department of Sleep Medicine Center, The Shengjing Affiliated Hospital,
China Medical University, Shenyang 110000, Liaoning, China
| | - Jinwei Li
- Department of Neurology/Stroke Center, the First Affiliated Hospital ofChina Medical University, China Medical University, Shenyang 110000, Liaoning, China
| | - Hongxi Li
- Department of Pain Management,
Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Yu Chen
- Department of The Fourth Otolaryngology Head and Neck Surgery,
Shengjing Hospital of China Medical University, Shenyang 110000, China
| |
Collapse
|
31
|
Liu Y, He Y, Xu H, Remmo A, Wiekhorst F, Heymann F, Liu H, Schellenberger E, Häckel A, Hauptmann R, Taupitz M, Shen Y, Yilmaz EY, Müller DN, Heidemann L, Schmidt R, Savic LJ. The Role of Glycocalyx Diversity and Thickness for Nanoparticle Internalization in M1-/M2-like Macrophages. NANO LETTERS 2024; 24:15607-15614. [PMID: 39621943 PMCID: PMC11638944 DOI: 10.1021/acs.nanolett.4c04004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024]
Abstract
Very small superparamagnetic iron oxide nanoparticles (VSOPs) show diagnostic value in multiple diseases as a promising MRI contrast agent. Macrophages predominantly ingest VSOPs, but the mechanism remains unclear. This study identifies differences in VSOP uptake between pro-inflammatory M1 and anti-inflammatory M2 macrophages and explores the role of the pericellular glycocalyx. Glycosaminoglycans (GAG) synthesis activities and the pericellular glycocalyx for M1/M2-like macrophages were assessed by RT-qPCR, Click-iT reaction, and WGA-FITC staining. The uptake of europium-VSOP and Synomag by the two subtypes was measured using Prussian blue staining, fluorescent microscopy, and magnetic particle spectroscopy. The findings revealed that M2-like macrophages had higher GAG synthesis activity, a thicker glycocalyx, and increased nanoparticle uptake compared to M1-like macrophages. Enzymatic glycocalyx degradation significantly decreased nanoparticle uptake. This study demonstrates a positive correlation between glycocalyx and nanoparticle uptake that could be exploited for imaging and targeted therapy, particularly in cancer, where macrophage subtypes play distinct roles.
Collapse
Affiliation(s)
- Yu Liu
- Department
of Radiology, Campus Virchow-Klinikum (CVK), Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
- Experimental
and Clinical Research Center, a joint cooperation
of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin
Berlin, Berlin 13125, Germany
| | - Yubei He
- Department
of Radiology, Campus Virchow-Klinikum (CVK), Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
- Experimental
and Clinical Research Center, a joint cooperation
of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin
Berlin, Berlin 13125, Germany
| | - Han Xu
- Department
of Radiology, Campus Virchow-Klinikum (CVK), Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
- Experimental
and Clinical Research Center, a joint cooperation
of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin
Berlin, Berlin 13125, Germany
| | - Amani Remmo
- Physikalisch-Technische
Bundesanstalt, Berlin 10587, Germany
| | - Frank Wiekhorst
- Physikalisch-Technische
Bundesanstalt, Berlin 10587, Germany
| | - Felix Heymann
- Department
of Hepatology, Campus Virchow-Klinikum (CVK), Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Hanyang Liu
- Department
of Hepatology, Campus Virchow-Klinikum (CVK), Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Eyk Schellenberger
- Department
of Radiology, Campus Charité Mitte (CCM), Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Akvile Häckel
- Department
of Radiology, Campus Charité Mitte (CCM), Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Ralf Hauptmann
- Department
of Radiology, Campus Charité Mitte (CCM), Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Matthias Taupitz
- Department
of Radiology, Campus Benjamin Franklin (CBF), Charité-Universitätsmedizin Berlin, Berlin 12203, Germany
| | - Yu Shen
- Deutsches
Rheuma-Forschungszentrum (DRFZ), Berlin 10117, Germany
| | - Emine Yaren Yilmaz
- Department
of Radiology, Campus Virchow-Klinikum (CVK), Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
- Experimental
and Clinical Research Center, a joint cooperation
of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin
Berlin, Berlin 13125, Germany
| | - Dominik N. Müller
- Experimental
and Clinical Research Center, a joint cooperation
of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin
Berlin, Berlin 13125, Germany
- Max
Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
- Charité-Universitätsmedizin
Berlin, Berlin 13125, Germany
| | - Luisa Heidemann
- Department
of Radiology, Campus Virchow-Klinikum (CVK), Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
- Experimental
and Clinical Research Center, a joint cooperation
of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin
Berlin, Berlin 13125, Germany
| | - Robin Schmidt
- Department
of Radiology, Campus Virchow-Klinikum (CVK), Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
- Experimental
and Clinical Research Center, a joint cooperation
of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin
Berlin, Berlin 13125, Germany
| | - Lynn Jeanette Savic
- Department
of Radiology, Campus Virchow-Klinikum (CVK), Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
- Experimental
and Clinical Research Center, a joint cooperation
of Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin
Berlin, Berlin 13125, Germany
- Berlin
Institute of Health at Charité-Universitätsmedizin
Berlin, Berlin 10178, Germany
| |
Collapse
|
32
|
Qin YS, Yi J, Chen YJ, Zhang W, Tang SF. Recent Advances in Micro/Nanomotor for the Therapy and Diagnosis of Atherosclerosis. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39648908 DOI: 10.1021/acsami.4c15165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Atherosclerotic cardiovascular disease poses a significant global public health threat with a high incidence that can result in severe mortality and disability. The lack of targeted effects from traditional therapeutic drugs on atherosclerosis may cause damage to other organs and tissues, necessitating the need for a more focused approach to address this dilemma. Micro/nanomotors are self-propelled micro/nanoscale devices capable of converting external energy into autonomous movement, which offers advantages in enhancing penetration depth and retention while increasing contact area with abnormal sites, such as atherosclerotic plaque, inflammation, and thrombosis, within blood vessel walls. Recent studies have demonstrated the crucial role micro/nanomotors play in treating atherosclerotic cardiovascular disease. Hence, this review highlights the recent progress of micro/nanomotor technology in atherosclerotic cardiovascular disease, including the effective promotion of micro/nanomotors in the circulatory system, overcoming hemorheological barriers, targeting the atherosclerotic plaque microenvironment, and targeting intracellular drug delivery, to facilitate atherosclerotic plaque localization and therapy. Furthermore, we also describe the potential application of micro/nanomotors in the imaging of vulnerable plaque. Finally, we discuss key challenges and prospects for treating atherosclerotic cardiovascular disease while emphasizing the importance of designing individualized management strategies specific to its causes and microenvironmental factors.
Collapse
Affiliation(s)
- Yu-Sheng Qin
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| | - Juan Yi
- Department of Laboratory Medicine, Liuzhou Traditional Chinese Medical Hospital, The Third Affiliated Hospital of Guangxi University of Chinese Medicine, Liuzhou 545006, China
| | - Yan-Jun Chen
- Department of Pathology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wei Zhang
- Department of Radiology, Liuzhou People's Hospital, Liuzhou 545006, China
| | - Shi-Fu Tang
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| |
Collapse
|
33
|
Gao H, Li J, Huang J, Jiang X. Screening and regulatory mechanism exploration of M1 macrophage polarization and efferocytosis-related biomarkers in coronary heart disease. Front Cardiovasc Med 2024; 11:1478827. [PMID: 39723414 PMCID: PMC11669322 DOI: 10.3389/fcvm.2024.1478827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
Background Macrophage polarization and efferocytosis have been implicated in CHD. However, the underlying mechanisms remain elusive. This study aimed to identify CHD-associated biomarkers using transcriptomic data. Methods This study examined 74 efferocytosis-related genes (ERGs) and 17 M1 macrophage polarization-related genes (MRGs) across two CHD-relevant datasets, GSE113079 and GSE42148. Differential expression analysis was performed separately on each dataset to identify differentially expressed genes (DEGs1 and DEGs2). The intersection of upregulated and downregulated genes from both sets was then used to define the final DEGs. Subsequently, MRG and ERG scores were calculated within the GSE113079 dataset, followed by weighted gene co-expression network analysis (WGCNA) to identify key module genes. The overlap between these module genes and the DEGs yielded candidate biomarkers, which were further evaluated through machine learning, receiver operating characteristic (ROC) curve analysis, and expression profiling. These biomarkers were subsequently leveraged to explore immune infiltration patterns and to construct a molecular regulatory network. To further validate their expression, quantitative reverse transcriptase PCR (qRT-PCR) was performed on clinical CHD samples, confirming the relevance and expression patterns of these biomarkers in the disease. Results A total of 93 DEGs were identified by intersecting the upregulated and downregulated genes from DEGs1 and DEGs2. WGCNA of the MRG and ERG scores identified 15,936 key module genes in the GSE113079 dataset. Machine learning and ROC analysis highlighted four biomarkers: C5orf58, CTAG1A, ZNF180, and IL13RA1. Among these, C5orf58, and ZNF180 were downregulated in CHD cases, while CTAG1A and IL13RA1 was upregulated. qRT-PCR results validated these findings for C5orf58, CTAG1A, ZNF180, and IL13RA1 showed inconsistent expression trends. Immune infiltration analysis indicated IL13RA1 all had a positive correlation with M0 macrophage, while had a negative correlation with. NK cells activated. The molecular regulatory network displayed that GATA2 and YY1 could regulate CTAG1A and ZNF180. Conclusions These results suggest that C5orf58, CTAG1A, ZNF180, and IL13RA1 serve as biomarkers linking M1 macrophage polarization and efferocytosis to CHD, providing valuable insights for CHD diagnosis and therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Xiaojie Jiang
- Department of Cardiology, The First Hospital of Nanchang, Nanchang, China
| |
Collapse
|
34
|
Liu Z, Yan F, Zhang H, Wang L, Zhao Y, Zhao H, Li C, Dai J, Yu B, Xiong H, Zhang J. Zingerone attenuates concanavalin A-induced acute liver injury by restricting inflammatory responses. Int Immunopharmacol 2024; 142:113198. [PMID: 39305891 DOI: 10.1016/j.intimp.2024.113198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 10/12/2024]
Abstract
Autoimmune hepatitis (AIH), an immune-mediated liver injury, plays an important role in the development and pathogenesis of several liver diseases. However, therapeutic alternatives for the treatment of AIH remain limited. Zingerone (ZIN) is a natural non-toxic phenolic compound extracted from ginger that possesses various pharmacological activities. Thus, this study aimed to investigate the effect of ZIN on AIH using a mouse model of acute liver injury induced by concanavalin A (Con A). To establish liver injury, C57BL/6J mice were intraperitoneally administered ZIN, followed by 20 mg/kg Con A after 3 h. Thereafter, the liver and serum were collected for analysis. The results revealed that ZIN pretreatment significantly suppressed the elevation of liver injury markers induced by Con A exposure and improved the survival of mice. Additionally, ZIN significantly ameliorated liver histopathological injury, hepatocyte apoptosis, and oxidative stress. Notably, ZIN inhibited hepatic M1 macrophage polarization and decreased the expression of M1 macrophage-associated pro-inflammatory genes and cytokines, including interleukin-1β (IL-1β), IL-12, IL-6, and tumor necrosis factor-α (TNF-α). Western blotting analysis indicated that ZIN inhibited the phosphorylation of extracellular receptor kin, c-Jun N-terminal kinase, and p65 in vitro. Taken together, these results suggest that ZIN exerts a protective effect in the Con A-induced acute liver injury model by inhibiting M1 macrophage polarization and suppressing NF-κB, mitogen-activated protein kinase, and interferon regulatory factor signaling pathways. This highlights the possibility of using ZIN as a safe drug for the treatment of liver injury and provides a novel therapeutic direction for clinical studies on liver diseases.
Collapse
Affiliation(s)
- Zhihong Liu
- School of Basic Medicine, Shandong First Medical University, Jinan 271016, China; Institute of Immunology and Molecular Medicine, Jining Medical University, Jining 272067, China
| | - Fenglian Yan
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining 272067, China; Jining Key Laboratory of Immunology, Jining Medical University, Jining 272067, China
| | - Hui Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining 272067, China; Jining Key Laboratory of Immunology, Jining Medical University, Jining 272067, China
| | - Lin Wang
- Cheeloo College of Medicine, Shandong University, Jinan 250000, Shandong, China
| | - Yuxuan Zhao
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining 272067, China; Jining Key Laboratory of Immunology, Jining Medical University, Jining 272067, China
| | - Hongru Zhao
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining 272067, China; Jining Key Laboratory of Immunology, Jining Medical University, Jining 272067, China
| | - Chunxia Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining 272067, China; Jining Key Laboratory of Immunology, Jining Medical University, Jining 272067, China
| | - Jun Dai
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining 272067, China; Jining Key Laboratory of Immunology, Jining Medical University, Jining 272067, China
| | - Bin Yu
- College of Integrated Chinese and Western Medicine, Jining Medical University, Jining 272067, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining 272067, China.
| | - Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining 272067, China.
| |
Collapse
|
35
|
Xiao Q, Huang J, Zhu X, Shi M, Chen L, Chen L, Liu X, Liu R, Zhong Y. Formononetin ameliorates dextran sulfate sodium-induced colitis via enhancing antioxidant capacity, promoting tight junction protein expression and reshaping M1/M2 macrophage polarization balance. Int Immunopharmacol 2024; 142:113174. [PMID: 39288627 DOI: 10.1016/j.intimp.2024.113174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/30/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
Ulcerative colitis (UC) is a complex, refractory inflammatory bowel disease characterized impared intestinal mucosal barrier and imbalanced M1/M2 macrophage polarization mediating its progression. Formononetin (FN), a bioactive isoflavone with established anti-inflammatory and immunomodulatory properties, shows promise in mitigating UC, yet its therapeutic and underlying mechanisms remain unclear. In this study, colitis was induced in mice by administering 2.5% (w/v) dextran sulfate sodium (DSS) solution for 7 days. Oral (25, 50, and 100 mg/kg) FN for 10 days significantly ameliorated colitis symptoms in a dose-dependent manner, by mitigating body weight loss, reducing disease activity index (DAI), colonic weight, and colonic weight index, while enhancing survival rates and colonic length. Histological analysis revealed FN remarkably suppressed inflammatory damage in colonic tissues. Furthermore, FN modulated the expression of pro- and anti-inflammatory cytokines and enhanced antioxidant capacity. Notably, FN treatment significantly enhanced the expression of tight junction (TJ) proteins (claudin-1, ZO-1, occludin) at both protein and mRNA levels in the colon tissues, suggesting improved intestinal barrier function. Crucially, FN inhibited macrophage infiltration in colonic tissues and rebalanced M1/M2 macrophage polarization. While, macrophage depletion largely abrogated FN's protective effects against colitis, indicating a crucial role for macrophages in mediating FN's therapeutic response. Overall, FN effectively alleviated colitis primarily via modulating inflammatory cytokine expression, enhancing antioxidant capacity, upregulating TJs proteins expression, and remodeling M1/M2 macrophage polarization equilibrium. These findings suggest that FN could be the next candidate to unlocking UC's treatment challenge.
Collapse
Affiliation(s)
- Qiuping Xiao
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Jiaqi Huang
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China; College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Xiyan Zhu
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China; College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Min Shi
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Liling Chen
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Lai Chen
- Institute of Cancer Research, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Xuan Liu
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Ronghua Liu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China.
| | - Youbao Zhong
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China; College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China.
| |
Collapse
|
36
|
Dalpati N, Rai SK, Sharma P, Sarangi PP. Integrins and Integrin-Driven Secretory Pathways as Multi-dimensional Regulators of Tumor-Associated Macrophage Recruitment and Reprogramming in Tumor Microenvironment. Matrix Biol 2024:S0945-053X(24)00145-8. [PMID: 39645091 DOI: 10.1016/j.matbio.2024.12.003] [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: 06/30/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Integrins, a group of transmembrane receptors, play a crucial role in mediating the interactions between cells and extracellular matrix (ECM) proteins. The intracellular signaling initiated by these cell-matrix interactions in leukocytes mediates many essential cellular processes such as survival, migration, metabolism, and other immunological functions. Macrophages, as phagocytes, participate in both proinflammatory and anti-inflammatory processes, including progression. Numerous reports have shown that the integrin-regulated secretome, comprising cytokines, chemokines, growth factors, proteases, and other bioactive molecules, is a crucial modulator of macrophage functions in tumors, significantly influencing macrophage programming and reprogramming within the tumor microenvironment (TME) in addition to driving their step-by-step entry process into tumor tissue spaces. Importantly, studies have demonstrated a pivotal role for integrin receptor-mediated secretome and associated signaling pathways in functional reprogramming from anti-tumorigenic to pro-tumorigenic phenotype in tumor-associated macrophages (TAMs). In this comprehensive review, we have provided an in-depth analysis of the latest findings of various key pathways, mediators, and signaling cascades associated with integrin-driven polarization of macrophages in tumors. This manuscript will provide an updated understanding of the modulation of inflammatory monocytes/ macrophages and TAMs by integrin-driven secretory pathways in various functions such as migration, differentiation, and their role in tumor progression, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Nibedita Dalpati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Shubham Kumar Rai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Prerna Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Pranita P Sarangi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.
| |
Collapse
|
37
|
Arachchi UPE, Madushani KP, Shanaka KASN, Kim G, Lim C, Yang H, Jayamali BPMV, Kodagoda YK, Warnakula WADLR, Jung S, Wan Q, Lee J. Characterization of tripartite motif containing 59 (TRIM59) in Epinephelus akaara: Insights into its immune involvement and functional properties in viral pathogenesis, macrophage polarization, and apoptosis regulation. FISH & SHELLFISH IMMUNOLOGY 2024; 157:110082. [PMID: 39645217 DOI: 10.1016/j.fsi.2024.110082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
The tripartite motif-containing (TRIM) superfamily is the largest family of RING-type E3 ubiquitin ligases that is conserved across the metazoan kingdom. Previous studies in mammals have demonstrated that TRIM59 possesses ubiquitin-protein ligase activity and acts as a negative regulator of NF-κB signaling. However, TRIM59 has rarely been characterized in fish. This study aimed to characterize TRIM59 from Epinephelus akaara (Eatrim59) and elucidate its structural features, expression patterns, and functional properties in innate immune responses and in the regulation of apoptosis. Eatrim59 is composed of 406 amino acids with a molecular weight of 45.84 kDa and a theoretical isoelectric point of 5.25. It comprises a conserved RING domain, a B-box motif, and a coiled-coil region. Subcellular localization analysis revealed that Eatrim59 was localized in the endoplasmic reticulum. Eatrim59 was ubiquitously expressed in all tissues examined, with the highest relative expression detected in the blood, followed by the brain and spleen. Temporal expression of Eatrim59 was dynamically regulated in response to in vivo immune stimulation by Toll-like receptor ligands and nervous necrosis virus infection. In FHM cells overexpressing Eatrim59, an increase in viral replication was observed upon infection with the Viral hemorrhagic septicemia virus. This phenomenon is attributed to Eatrim59-mediated downregulation of interferon, pro-inflammatory cytokines, and other antiviral pathways. Moreover, macrophages stably overexpressing Eatrim59 exhibited a decrease in nitric oxide production and the formation of a filamentous actin structure upon lipopolysaccharide stimulation, indicating dampened M1 polarization. Furthermore, a decrease in apoptosis was observed in Eatrim59-overexpressing FHM cells under oxidative stress induced by H2O2. In conclusion, these findings demonstrate the multifaceted role of Eatrim59 as a regulator of innate immune response and apoptosis in E. akaara.
Collapse
Affiliation(s)
- U P E Arachchi
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - K P Madushani
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - K A S N Shanaka
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Gaeun Kim
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Chaehyeon Lim
- Genetics and Breeding Research Center, National Institute of Fisheries Science, Geoje, 53334, Republic of Korea
| | - Hyerim Yang
- Genetics and Breeding Research Center, National Institute of Fisheries Science, Geoje, 53334, Republic of Korea
| | - B P M Vileka Jayamali
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Yasara Kavindi Kodagoda
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - W A D L R Warnakula
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Sumi Jung
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - Qiang Wan
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Jeju National University, Jeju, 63333, Republic of Korea.
| |
Collapse
|
38
|
Yang Y, Xie T, Gao P, Han W, Liu Y, Wang Y. Hsa_Circ_002144 Promotes Glycolysis and Immune Escape of Breast Cancer Through miR-326/PKM Axis. Cancer Biother Radiopharm 2024; 39:755-769. [PMID: 38963787 DOI: 10.1089/cbr.2024.0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024] Open
Abstract
Background: Breast cancer is a leading cause of cancer-related deaths in women worldwide, posing a significant threat to female health. Therefore, it is crucial to search for new therapeutic targets and prognostic biomarkers for breast cancer patients. Method: Bioinformatics analysis, quantitative real-time PCR (qRT-PCR), and fluorescence in situ hybridization (FISH) were employed to investigate the expression of hsa_circ_002144 in breast cancer. Transwell assay, Western blotting, and cell viability assay were utilized to assess the impact of hsa_circ_002144 on the proliferation, migration, and invasion of breast cancer cells. Additionally, a mouse model was established to validate its functionality. Flow cytometry, WB analysis, enzyme-linked immunosorbent assay (ELISA), qRT-PCR, exosomes isolation, and co-culture system were employed to elucidate the molecular mechanism underlying macrophage polarization. Result: we have discovered for the first time that hsa_circ_002144 is highly expressed in breast cancer. It affected tumor growth and metastasis and could influence macrophage polarization through the glycolytic pathway. Conclusion: This finding provides a new direction for breast cancer treatment and prognosis assessment.
Collapse
Affiliation(s)
- Yong Yang
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang City, China
| | - Tianhao Xie
- General Surgery, The Affiliated Hospital of Hebei University, Baoding City, China
| | - Peng Gao
- Anesthesiology department, Affiliated hospital of Qingdao university, Qingdao City, China
| | - Weijun Han
- Third Surgery, Baoji traditional Chinese Medicine Hospital in Shaanxi Province, Baoji City, China
| | - Yuhong Liu
- Rheumatology and Immunology Department, The Affiliated Hospital of Yan 'an University, Yan 'an City, China
| | - Yanmei Wang
- School of Nursing and Health, Medical College of Yan 'an University, Yan 'an City, China
| |
Collapse
|
39
|
Feng X, Lai X, Zhou M, Bie J, Li T, Wang D, Chen S, Hu X, Wang C, Xu P. Targeting HLA-E in Lung Cancer: The Therapeutic Potential of IRF5-Engineered M1-Macrophage-Derived Exosomes. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e70035. [PMID: 39623605 PMCID: PMC11611755 DOI: 10.1111/crj.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/23/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024]
Abstract
Immunotherapy is a pivotal approach in the treatment of lung cancer. Although HLA-E is a potential target for tumor immunotherapy, its role in lung cancer remains unclear. Previous studies have identified the transcription factor IRF5 as a characteristic gene of M1-like macrophages, highlighting its crucial role in promoting antitumor immune responses. In this study, we developed an engineered M1-like macrophage exosomes expressing IRF5 (IRF5 M1-exos) and demonstrated their ability to inhibit proliferation, migration, and invasion of lung cancer cells. Moreover, our experiments using a nude mouse model revealed that IRF5 M1-exos exerted potent therapeutic effects by effectively suppressing tumor growth. Notably, the mechanism by which IRF5 exerts its antitumor function through HLA-E regulation in lung cancer has not been fully elucidated. Here, we identified HLA-E as a downstream target gene of IRF5 and demonstrated that the overexpression of HLA-E can counteract the tumor-promoting effects induced by si-IRF5 M1-exos. These results suggest that M1 macrophage-derived exosomes, enriched with the transcription factor IRF5, exhibit potent antitumor activity by up-regulating HLA-E in lung cancer cells. Therefore, IRF5 M1-exos represent an attractive therapeutic strategy for lung cancer.
Collapse
Affiliation(s)
- Xuqin Feng
- Department of OncologyBeijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), The Second Clinical Medical College of North Sichuan Medical CollegeNanchongSichuanChina
| | - Xiangyu Lai
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing Cancer HospitalChongqing Cancer InstituteChongqingChina
| | - Mingming Zhou
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing Cancer HospitalChongqing Cancer InstituteChongqingChina
| | - Jun Bie
- Department of OncologyBeijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), The Second Clinical Medical College of North Sichuan Medical CollegeNanchongSichuanChina
| | - Tingting Li
- Department of OncologyBeijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), The Second Clinical Medical College of North Sichuan Medical CollegeNanchongSichuanChina
| | - Dan Wang
- Department of OncologyBeijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), The Second Clinical Medical College of North Sichuan Medical CollegeNanchongSichuanChina
| | - Silin Chen
- Department of OncologyBeijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), The Second Clinical Medical College of North Sichuan Medical CollegeNanchongSichuanChina
| | - Xin Hu
- Department of OncologyBeijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), The Second Clinical Medical College of North Sichuan Medical CollegeNanchongSichuanChina
| | - Chunyu Wang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing Cancer HospitalChongqing Cancer InstituteChongqingChina
| | - Peng Xu
- Department of Clinical LaboratoryBeibei Traditional Chinese Medical HospitalChongqingChina
| |
Collapse
|
40
|
Luo L, Zhuang X, Fu L, Dong Z, Yi S, Wang K, Jiang Y, Zhao J, Yang X, Hei F. The role of the interplay between macrophage glycolytic reprogramming and NLRP3 inflammasome activation in acute lung injury/acute respiratory distress syndrome. Clin Transl Med 2024; 14:e70098. [PMID: 39623879 PMCID: PMC11612265 DOI: 10.1002/ctm2.70098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/26/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a severe respiratory condition associated with elevated morbidity and mortality. Understanding their complex pathophysiological mechanisms is crucial for developing new preventive and therapeutic strategies. Recent studies highlight the significant role of inflammation involved in ALI/ARDS, particularly the hyperactivation of the NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome in macrophages. This activation drives pulmonary inflammation by releasing inflammatory signalling molecules and is linked to metabolic reprogramming, marked by increased glycolysis and reduced oxidative phosphorylation. However, the relationship between NLRP3 inflammasome activation and macrophage glycolytic reprogramming in ALI/ARDS, as well as the molecular mechanisms regulating these processes, remain elusive. This review provides a detailed description of the interactions and potential mechanisms linking NLRP3 inflammasome activation with macrophage glycolytic reprogramming, proposing that glycolytic reprogramming may represent a promising therapeutic target for mitigating inflammatory responses in ALI/ARDS. KEY POINTS: NLRP3 inflammasome activation is pivotal in mediating the excessive inflammatory response in ALI/ARDS. Glycolytic reprogramming regulates NLRP3 inflammasome activation. Therapeutic potential of targeting glycolytic reprogramming to inhibit NLRP3 inflammasome activation in ALI/ARDS.
Collapse
Affiliation(s)
- Lan Luo
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Xiaoli Zhuang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Lin Fu
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Ziyuan Dong
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Shuyuan Yi
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Kan Wang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yu Jiang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Ju Zhao
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Xiaofang Yang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Feilong Hei
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
41
|
Wu M, Fletcher EL, Chinnery HR, Downie LE, Mueller SN. Redefining our vision: an updated guide to the ocular immune system. Nat Rev Immunol 2024; 24:896-911. [PMID: 39215057 DOI: 10.1038/s41577-024-01064-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2024] [Indexed: 09/04/2024]
Abstract
Balanced immune responses in the eyes are crucial to preserve vision. The ocular immune system has long been considered distinct, owing to the so-called 'immune privilege' of its component tissues. More recently, intravital imaging and transcriptomic techniques have reshaped scientific understanding of the ocular immune landscape, such as revealing the specialization of immune cell populations in the various tissues of the eye. As knowledge of the phenotypes of corneal and retinal immune cells has evolved, links to both the systemic immune system, and the central and peripheral nervous systems, have been identified. Using intravital imaging, T cells have recently been found to reside in, and actively patrol, the healthy human cornea. Disease-associated retinal microglia with links to retinal degeneration have also been identified. This Review provides an updated guide to the ocular immune system, highlighting current knowledge of the immune cells that are present in steady-state and specific diseased ocular tissues, as well as evidence for their relationship to systemic disease. In addition, we discuss emerging intravital imaging techniques that can be used to visualize immune cell morphology and dynamics in living human eyes and how these could be applied to advance understanding of the human immune system.
Collapse
Affiliation(s)
- Mengliang Wu
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Erica L Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Carlton, Victoria, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia.
- Lions Eye Institute, Nedlands, Western Australia, Australia.
- Optometry, The University of Western Australia, Crawley, Western Australia, Australia.
| | - Laura E Downie
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia.
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
42
|
Wang X, Liu D. Macrophage Polarization: A Novel Target and Strategy for Pathological Scarring. Tissue Eng Regen Med 2024; 21:1109-1124. [PMID: 39352458 PMCID: PMC11589044 DOI: 10.1007/s13770-024-00669-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Abnormal scarring imposes considerable challenges and burdens on the lives of patients and healthcare system. Macrophages at the wound site are found to be of great concern to overall wound healing. There have been many studies indicating an inextricably link between dysfunctional macrophages and fibrotic scars. Macrophages are not only related to pathogen destruction and phagocytosis of apoptotic cells, but also involved in angiogenesis, keratinization and collagen deposition. These abundant cell functions are attributed to specific heterogeneity and plasticity of macrophages, which also add an extra layer of complexity to correlational researches. METHODS This article summarizes current understanding of macrophage polarization in scar formation and several prevention and treatment strategies on pathological scarring related to regulation of macrophage behaviors by utilizing databases such as PubMed, Google Scholar and so on. RESULTS There are many studies proving that macrophages participate in the course of wound healing by converting their predominant phenotype. The potential of macrophages in managing hypertrophic scars and keloid lesions have been underscored. CONCLUSION Macrophage polarization offers new prevention strategies for pathological scarring. Learning about and targeting at macrophages may be helpful in achieving optimum wound healing.
Collapse
Affiliation(s)
- Xinyi Wang
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
- Queen Mary Academy, Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Dewu Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
43
|
Kzhyshkowska J, Shen J, Larionova I. Targeting of TAMs: can we be more clever than cancer cells? Cell Mol Immunol 2024; 21:1376-1409. [PMID: 39516356 DOI: 10.1038/s41423-024-01232-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
АBSTRACT: With increasing incidence and geography, cancer is one of the leading causes of death, reduced quality of life and disability worldwide. Principal progress in the development of new anticancer therapies, in improving the efficiency of immunotherapeutic tools, and in the personification of conventional therapies needs to consider cancer-specific and patient-specific programming of innate immunity. Intratumoral TAMs and their precursors, resident macrophages and monocytes, are principal regulators of tumor progression and therapy resistance. Our review summarizes the accumulated evidence for the subpopulations of TAMs and their increasing number of biomarkers, indicating their predictive value for the clinical parameters of carcinogenesis and therapy resistance, with a focus on solid cancers of non-infectious etiology. We present the state-of-the-art knowledge about the tumor-supporting functions of TAMs at all stages of tumor progression and highlight biomarkers, recently identified by single-cell and spatial analytical methods, that discriminate between tumor-promoting and tumor-inhibiting TAMs, where both subtypes express a combination of prototype M1 and M2 genes. Our review focuses on novel mechanisms involved in the crosstalk among epigenetic, signaling, transcriptional and metabolic pathways in TAMs. Particular attention has been given to the recently identified link between cancer cell metabolism and the epigenetic programming of TAMs by histone lactylation, which can be responsible for the unlimited protumoral programming of TAMs. Finally, we explain how TAMs interfere with currently used anticancer therapeutics and summarize the most advanced data from clinical trials, which we divide into four categories: inhibition of TAM survival and differentiation, inhibition of monocyte/TAM recruitment into tumors, functional reprogramming of TAMs, and genetic enhancement of macrophages.
Collapse
Affiliation(s)
- Julia Kzhyshkowska
- Department of Innate Immunity and Tolerance, Institute of Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer, 1-3, 68167, Mannheim, Germany.
- German Red Cross Blood Service Baden-Württemberg - Hessen, Friedrich-Ebert Str. 107, 68167, Mannheim, Germany.
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050, Lenina av.36, Tomsk, Russia.
- Bashkir State Medical University of the Ministry of Health of Russia, 450000, Teatralnaya Street, 2a, Ufa, Russia.
| | - Jiaxin Shen
- Department of Innate Immunity and Tolerance, Institute of Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer, 1-3, 68167, Mannheim, Germany
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050, Lenina av.36, Tomsk, Russia
- Bashkir State Medical University of the Ministry of Health of Russia, 450000, Teatralnaya Street, 2a, Ufa, Russia
- Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009, Kooperativnyi st, Tomsk, Russia
| |
Collapse
|
44
|
Zhang Y, Dai J, Hang R, Yao X, Bai L, Wang H, Huang D, Hang R. Tailoring surface stiffness to modulate senescent macrophage immunomodulation: Implications for osteo-/angio-genesis in aged bone regeneration. BIOMATERIALS ADVANCES 2024; 165:214010. [PMID: 39222592 DOI: 10.1016/j.bioadv.2024.214010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/09/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
The application of biomaterials in bone regeneration is a prevalent clinical practice. However, its efficacy in elderly patients remains suboptimal, necessitating further advancements. While biomaterial properties are known to orchestrate macrophage (MΦ) polarization and local immune responses, the role of biomaterial cues, specifically stiffness, in directing the senescent macrophage (S-MΦ) is still poorly understood. This study aimed to elucidate the role of substrate stiffness in modulating the immunomodulatory properties of S-MΦ and their role in osteo-immunomodulation. Our results demonstrated that employing collagen-coated polyacrylamide hydrogels with varying stiffness values (18, 76, and 295 kPa) as model materials, the high-stiffness hydrogel (295 kPa) steered S-MΦs towards a pro-inflammatory M1 phenotype, while hydrogels with lower stiffness (18 and 76 kPa) promoted an anti-inflammatory M2 phenotype. The immune microenvironment created by S-MΦs promoted the bioactivities of senescent endothelial cells (S-ECs) and senescent bone marrow mesenchymal stem cells BMSCs (S-BMSCs). Furthermore, the M2 S-MΦs, particularly incubated on the 76 kPa hydrogel matrices, significantly enhanced the ability of angiogenesis of S-ECs and osteogenic differentiation of S-BMSCs, which are crucial and interrelated processes in bone healing. This modulation aided in reducing the accumulation of reactive oxygen species in S-ECs and S-BMSCs, thereby significantly contributing to the repair and regeneration of aged bone tissue.
Collapse
Affiliation(s)
- Yi Zhang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Jinjun Dai
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Ruiyue Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Xiaohong Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China.
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.
| | - Huaiyu Wang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology Chinese Academy of Sciences, Shenzhen 518055, China
| | - Di Huang
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030060, China
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China.
| |
Collapse
|
45
|
Jin S, Yu Y, Zhang T, Xie D, Zheng Y, Wang C, Liu Y, Xia D. Surface modification strategies to reinforce the soft tissue seal at transmucosal region of dental implants. Bioact Mater 2024; 42:404-432. [PMID: 39308548 PMCID: PMC11415887 DOI: 10.1016/j.bioactmat.2024.08.042] [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: 07/20/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Soft tissue seal around the transmucosal region of dental implants is crucial for shielding oral bacterial invasion and guaranteeing the long-term functioning of implants. Compared with the robust periodontal tissue barrier around a natural tooth, the peri-implant mucosa presents a lower bonding efficiency to the transmucosal region of dental implants, due to physiological structural differences. As such, the weaker soft tissue seal around the transmucosal region can be easily broken by oral pathogens, which may stimulate serious inflammatory responses and lead to the development of peri-implant mucositis. Without timely treatment, the curable peri-implant mucositis would evolve into irreversible peri-implantitis, finally causing the failure of implantation. Herein, this review has summarized current surface modification strategies for the transmucosal region of dental implants with improved soft tissue bonding capacities (e.g., improving surface wettability, fabricating micro/nano topographies, altering the surface chemical composition and constructing bioactive coatings). Furthermore, the surfaces with advanced soft tissue bonding abilities can be incorporated with antibacterial properties to prevent infections, and/or with immunomodulatory designs to facilitate the establishment of soft tissue seal. Finally, we proposed future research orientations for developing multifunctional surfaces, thus establishing a firm soft tissue seal at the transmucosal region and achieving the long-term predictability of dental implants.
Collapse
Affiliation(s)
- Siqi Jin
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Yameng Yu
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| | - Ting Zhang
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Daping Xie
- State Key Laboratory in Quality Research of Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, 999078, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
- International Research Organization for Advanced Science and Technology (IROAST), Kumamoto University, 2-39-2 Kumamoto, 860-8555, Japan
| | - Chunming Wang
- State Key Laboratory in Quality Research of Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, 999078, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Dandan Xia
- Department of Dental Materials, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, 100081, China
| |
Collapse
|
46
|
Zheng M, Li J, Cao Y, Bao Z, Dong X, Zhang P, Yan J, Liu Y, Guo Y, Zeng X. Association of different inflammatory indices with risk of early natural menopause: a cross-sectional analysis of the NHANES 2013-2018. Front Med (Lausanne) 2024; 11:1490194. [PMID: 39678034 PMCID: PMC11638831 DOI: 10.3389/fmed.2024.1490194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024] Open
Abstract
Background Early natural menopause, characterized by the cessation of ovarian function before the age of 45, has been a subject of prior research indicating that inflammation may predict the onset of menopause. However, the specific relationship between peripheral blood inflammatory parameters and early natural menopause remains ambiguous. Methods This observational study utilized data from the National Health and Nutrition Examination Survey (NHANES) spanning the years 2013-2018. The age at menopause was ascertained through the Reproductive Health Questionnaire (RHQ), with early natural menopause defined as menopause occurring before the age of 45 years. Complete blood counts were derived from laboratory test data, and seven indices of inflammation were calculated, including lymphocyte count (LC), neutrophil count (NC), systemic immune inflammation index (SII), product of platelet and neutrophil count (PPN), platelet-lymphocyte ratio (PLR), neutrophil-lymphocyte ratio (NLR), and lymphocyte-monocyte ratio (LMR). A multivariate weighted logistic regression analysis was employed to estimate the association between these inflammatory indices and early natural menopause. Results A total of 2,034 participants were included in the analysis, of whom 460 reported experiencing menopause before the age of 45. Both Log2-NC and Log2-PPN were found to be positively correlated with early menopause, with odds ratios (OR) of 1.56 (95% CI: 1.16, 2.09; p = 0.005) and 1.36 (95% CI: 1.07, 1.72; p = 0.015), respectively. The results from models 1 and 2 were consistent with those from model 3. In the trend test, participants in the fourth quartile (Q4) of log2-LC exhibited a positive correlation with early menopause compared to those in the lowest quartile (Q1), with an OR of 1.41 (95% CI: 1.03, 1.93; p = 0.033). Similarly, the fourth quartile (Q4) of log2-NC and log2-PPN demonstrated a positive correlation with early menopause, with odds ratios (OR) of 1.76 (95% CI: 1.27-2.45; p < 0.001) and 1.66 (95% CI: 1.21-2.29; p = 0.002), respectively. In Model 3, log2-SII, log2-PLR, log2-NLR, and log2-LMR were not significantly associated with early menopause. Conclusion Our findings indicate that elevated levels of Log2-LC, Log2-NC, and Log2-PPN are positively correlated with an increased risk of early menopause among women in the United States.
Collapse
Affiliation(s)
- Mengyu Zheng
- Department of Gynecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
| | - Junying Li
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yushan Cao
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhuo Bao
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xing Dong
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pei Zhang
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinxiang Yan
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yixuan Liu
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongzhen Guo
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xianxu Zeng
- Zhengzhou Key Laboratory of Gynecological Disease's Early Diagnosis, Zhengzhou, China
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
47
|
Tedeschi G, Navarro MX, Scipioni L, Sondhi TK, Prescher JA, Digman MA. Monitoring Macrophage Polarization with Gene Expression Reporters and Bioluminescence Phasor Analysis. CHEMICAL & BIOMEDICAL IMAGING 2024; 2:765-774. [PMID: 39610466 PMCID: PMC11600157 DOI: 10.1021/cbmi.4c00049] [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: 06/13/2024] [Revised: 08/06/2024] [Accepted: 08/19/2024] [Indexed: 11/30/2024]
Abstract
Macrophages exhibit a spectrum of behaviors upon activation and are generally classified as one of two types: inflammatory (M1) or anti-inflammatory (M2). Tracking these phenotypes in living cells can provide insight into immune function but remains a challenging pursuit. Existing methods are mostly limited to static readouts or are difficult to employ for multiplexed imaging in complex 3D environments while maintaining cellular resolution. We aimed to fill this void using bioluminescent technologies. Here we report genetically engineered luciferase reporters for the long-term monitoring of macrophage polarization via spectral phasor analysis. M1- and M2-specific promoters were used to drive the expression of bioluminescent enzymes in macrophage cell lines. The readouts were multiplexed and discernible in both 2D and 3D formats with single-cell resolution in living samples. Collectively, this work expands the toolbox of methods for monitoring macrophage polarization and provides a blueprint for monitoring other multifaceted networks in heterogeneous environments.
Collapse
Affiliation(s)
- Giulia Tedeschi
- Laboratory
for Fluorescence Dynamics, Biomedical Engineering Department, University of California, Irvine, Irvine, California 92617, United States
| | - Mariana X. Navarro
- Department
of Chemistry, University of California Irvine, Irvine, California 92617, United States
| | - Lorenzo Scipioni
- Laboratory
for Fluorescence Dynamics, Biomedical Engineering Department, University of California, Irvine, Irvine, California 92617, United States
| | - Tanvi K. Sondhi
- Department
of Chemistry, University of California Irvine, Irvine, California 92617, United States
| | - Jennifer A. Prescher
- Department
of Chemistry, University of California Irvine, Irvine, California 92617, United States
- Department
of Molecular Biology and Biochemistry, University
of California, Irvine, Irvine, California 92617, United States
- Department
of Pharmaceutical Sciences, University of
California, Irvine, Irvine, California 92617, United States
| | - Michelle A. Digman
- Laboratory
for Fluorescence Dynamics, Biomedical Engineering Department, University of California, Irvine, Irvine, California 92617, United States
| |
Collapse
|
48
|
Angarola BL, Sharma S, Katiyar N, Kang HG, Nehar-Belaid D, Park S, Gott R, Eryilmaz GN, LaBarge MA, Palucka K, Chuang JH, Korstanje R, Ucar D, Anczuków O. Comprehensive single-cell aging atlas of healthy mammary tissues reveals shared epigenomic and transcriptomic signatures of aging and cancer. NATURE AGING 2024:10.1038/s43587-024-00751-8. [PMID: 39587369 DOI: 10.1038/s43587-024-00751-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 10/16/2024] [Indexed: 11/27/2024]
Abstract
Aging is the greatest risk factor for breast cancer; however, how age-related cellular and molecular events impact cancer initiation is unknown. In this study, we investigated how aging rewires transcriptomic and epigenomic programs of mouse mammary glands at single-cell resolution, yielding a comprehensive resource for aging and cancer biology. Aged epithelial cells exhibit epigenetic and transcriptional changes in metabolic, pro-inflammatory and cancer-associated genes. Aged stromal cells downregulate fibroblast marker genes and upregulate markers of senescence and cancer-associated fibroblasts. Among immune cells, distinct T cell subsets (Gzmk+, memory CD4+, γδ) and M2-like macrophages expand with age. Spatial transcriptomics reveals co-localization of aged immune and epithelial cells in situ. Lastly, we found transcriptional signatures of aging mammary cells in human breast tumors, suggesting possible links between aging and cancer. Together, these data uncover that epithelial, immune and stromal cells shift in proportions and cell identity, potentially impacting cell plasticity, aged microenvironment and neoplasia risk.
Collapse
Affiliation(s)
| | | | - Neerja Katiyar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Hyeon Gu Kang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - SungHee Park
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Giray N Eryilmaz
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Mark A LaBarge
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA.
- Institute for Systems Genomics, UConn Health, Farmington, CT, USA.
| | - Olga Anczuków
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA.
- Institute for Systems Genomics, UConn Health, Farmington, CT, USA.
| |
Collapse
|
49
|
Petrosiute A, Musvicaitė J, Petroška D, Ščerbavičienė A, Arnold S, Matulienė J, Žvirblienė A, Matulis D, Lučiūnaitė A. CCL2-CCR2 Axis Inhibition in Osteosarcoma Cell Model: The Impact of Oxygen Level on Cell Phenotype. J Cell Physiol 2024. [PMID: 39587819 DOI: 10.1002/jcp.31489] [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: 05/24/2024] [Revised: 10/14/2024] [Accepted: 11/06/2024] [Indexed: 11/27/2024]
Abstract
Treatment of osteosarcoma is hampered by tumor hypoxia and requires alternative approaches. Although the CCL2-CCR2 axis is indispensable in tumor-induced inflammation and angiogenesis, its blockade has not been effective to date. This study aimed to characterize how CCR2 inhibition affects the crosstalk of osteosarcoma cells with immune cells to better delineate tumor resistance mechanisms that help withstand such treatment. In this study, 143B cells were exposed to healthy donor PBMC supernatants in a transwell assay lacking direct cell-to-cell contact and subjected to different oxygen concentrations. In addition, mice bearing orthotopic 143B tumors were subjected to CCR2 antagonist treatment. Our findings show that hypoxic conditions alter cytokine and cancer- related protein expression on cells and impair CCR2 antagonist effects in the experimental osteosarcoma model. CCL2-CCR2 axis blockade in the 143B xenografts, which are positive for hypoxia marker CAIX, did not slow 143B tumor growth or metastasis but altered tumor microenvironment by VEGFR downregulation and shift in the CD44-positive cell population towards high CD44 expression. This study highlights differential responses of tumor cells to CCR2 antagonists in the presence of different oxygen saturations and expands our knowledge of compensatory mechanisms leading to CCL2-CCR2 treatment resistance.
Collapse
Affiliation(s)
- Agne Petrosiute
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Justina Musvicaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Donatas Petroška
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Alvilė Ščerbavičienė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Sascha Arnold
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Jurgita Matulienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Aurelija Žvirblienė
- Department of Immunology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Asta Lučiūnaitė
- Department of Immunology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
50
|
Xu H, Ding S, Tong Y, Zhang Q. Genetic Evidence of Obesity-Induced Chronic Wounds Mediated by Inflammatory Biomarkers. Biol Res Nurs 2024:10998004241299375. [PMID: 39568230 DOI: 10.1177/10998004241299375] [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: 11/22/2024]
Abstract
Background: Obese patients are increasingly recognized as being at higher risk for skin diseases, particularly chronic wounds. While the exact mechanisms remain unclear, obesity is suspected to influence the development of chronic injuries via inflammatory biomarkers. Single nucleotide polymorphisms (SNPs) may further influence gene expression, protein function, and levels of inflammatory biomarkers through various mechanisms, thereby modulating inflammatory responses that contribute to wound pathogenesis. Methods: A two-sample two-step Mendelian Randomization (MR) was employed to explore the causal relationship between obesity and chronic wounds, focusing on the mediating role of inflammatory biomarkers. SNPs were used as instrumental variables (IVs) to infer causality. Obesity-related genetic data were sourced from the UK Biobank and GIANT consortium. Genome-wide association studies provided data on 92 inflammatory biomarkers, involving 14,824 and 575,531 individuals. Pressure injuries, lower limb venous ulcers, and diabetic foot ulcer data were obtained from FinnGen R10 and the Pan-UK Biobank. Results: Obesity significantly increased the risk of pressure injuries, lower limb venous ulcers, and diabetic foot ulcers. CCL19, hGDNF, IL-12B, and TNFRSF9 were identified as mediators in obesity-induced lower limb venous ulcers. Conclusion: This study provides genetic evidence that obesity leads to lower limb venous ulcers via inflammatory biomarkers, suggesting potential therapeutic targets for intervention.
Collapse
Affiliation(s)
- Hai Xu
- Nursing College, Hangzhou Normal University, Hangzhou, China
- Department of Nursing, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Sheyuan Ding
- Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou, China
- The Affiliated Hospital of Hangzhou Normal University (School of Clinical Medicine, School of Stomatology), Hangzhou, China
| | - Yu Tong
- Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Qiong Zhang
- Nursing College, Hangzhou Normal University, Hangzhou, China
- Department of Nursing, Zhejiang Provincial People's Hospital, Hangzhou, China
- Center for Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| |
Collapse
|