1
|
Yu W, Hu L, Wei Y, Xue C, Liu Y, Xie H. Advances of novel hydrogels in the healing process of alveolar sockets. BIOMATERIALS ADVANCES 2025; 173:214280. [PMID: 40086007 DOI: 10.1016/j.bioadv.2025.214280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Tooth extraction is a common oral surgical procedure that often leads to delayed alveolar socket healing due to the complexity of the oral microenvironment, which can hinder the patient's aesthetic and functional recovery. Effective alveolar socket healing requires a multidisciplinary approach. Recent advancements in materials science and bioengineering have facilitated the development of innovative strategies, with hydrogels emerging as ideal restorative materials for alveolar socket repair due to their superior properties. This review provides an overview of recent advances in hydrogels for alveolar socket healing, focusing on their classification, physical properties (e.g., mechanical strength, swelling behavior, degradation rate, and injectability), biological functions, and applications in relevant animal models. Specifically, the bone-regenerative and antimicrobial properties of hydrogels are highlighted. Furthermore, this review identifies future directions and addresses challenges associated with the clinical application of hydrogels in extraction socket healing.
Collapse
Affiliation(s)
- Wenqing Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Liwei Hu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Yige Wei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Chengyu Xue
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Yunfei Liu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing 401147, PR China.
| | - Huixu Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
2
|
Lu J, Zhou Y, Song YX, Wang JY, Xian JX. Natural alkaloids modulating macrophage polarization: Innovative therapeutic strategies for inflammatory, cardiovascular, and cancerous diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156709. [PMID: 40250001 DOI: 10.1016/j.phymed.2025.156709] [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: 12/31/2024] [Revised: 03/21/2025] [Accepted: 03/29/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Macrophage polarization, switching between pro-inflammatory M1 and anti-inflammatory M2 states, is crucial for disease dynamics in inflammatory, metabolic, and cancer contexts. Modulating this polarization is a clinical challenge, but natural alkaloids, with their potent anti-inflammatory and immunomodulatory effects, show promise in reprogramming macrophage phenotypes. PURPOSE This review explores the applications of natural alkaloids-such as matrine, berberine, koumine, sophoridine, and curcumin-in modulating macrophage polarization. It aims to highlight their potential in reprogramming macrophage phenotypes and improving therapeutic outcomes across various diseases. METHODS A comprehensive literature review was conducted using databases like PubMed, Web of Science, Science Direct and Google Scholar, employing targeted keywords related to natural alkaloids, macrophage polarization, and disease treatment. The analysis primarily focused on articles published between 2020 and 2024. RESULTS This review summarizes how natural alkaloids regulate macrophage polarization, promoting the M2 phenotype to reduce inflammation, thereby playing a therapeutic role in anti-inflammatory, cardiovascular, and metabolic diseases. At the same time, they also promote M1 polarization to inhibit tumor development. CONCLUSION Accumulating evidence demonstrates that macrophage polarization regulation by natural alkaloids holds notable clinical value for disease intervention. They alleviate inflammation, enhance antitumor immunity, and improve treatment outcomes, demonstrating their importance in innovative therapeutic strategies. Moreover, combining alkaloids with immunotherapy enhances treatment efficacy, further highlighting their versatility in a variety of therapeutic applications.
Collapse
Affiliation(s)
- Jing Lu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Ying Zhou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yi-Xuan Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jie-Ying Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jia-Xun Xian
- Traditional Chinese Medicine Hospital of Meishan, Meishan 620010, China.
| |
Collapse
|
3
|
Solarska-Ściuk K, Pruchnik H. A Critical View on the Biocompatibility of Silica Nanoparticles and Liposomes as Drug Delivery Systems. Mol Pharm 2025. [PMID: 40340409 DOI: 10.1021/acs.molpharmaceut.5c00501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Silica-based materials and liposomes are widely employed in drug delivery systems, particularly as the most frequently evaluated platforms for intravenous drug administration. Their exceptional biocompatibility, versatile surface modification capabilities, and efficient encapsulation of a broad spectrum of therapeutic agents make them ideal for targeted and controlled drug delivery. Both nanodelivery systems interact with endothelial cells and various blood components, including erythrocytes (red blood cells) and white blood cells (lymphocytes, monocytes, and macrophages), potentially leading to cytotoxic effects. However, the detrimental impacts of silica nanoparticles (MSNs) and liposomes on healthy cells remain insufficiently investigated. The cytotoxicity of these carriers is strongly influenced by their physicochemical properties, such as size, surface charge, and functionalization, as well as the specific type of cells they encounter. This review aims to explore the molecular and cellular dysfunctions induced by MSNs and liposomes, which elicit various biological responses, including proinflammatory signaling, oxidative stress, and autophagy. Considering the toxicity associated with nanosilica and liposomes, strategies such as surface modifications and morphological adjustments may serve as effective approaches to mitigate these adverse effects. Implementing such modifications holds the potential to develop nanomaterials with lower toxicological profiles, thereby enhancing their safety and efficacy in clinical applications. By addressing these challenges, the advancement of silica-based materials and liposomes can be optimized for safer and more effective intravenous drug delivery systems.
Collapse
Affiliation(s)
- Katarzyna Solarska-Ściuk
- Faculty of Biotechnology, Collegium Medicum, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Hanna Pruchnik
- Department of Physics and Biophysics, Faculty of Biotechnology and Food Sciences, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375 Wrocław, Poland
| |
Collapse
|
4
|
Hussen J, Al-Mubarak AIA, Shawaf T, Bukhari K, Alkharsah KR. Modulatory Effects of the Recombinant Middle East Respiratory Syndrome Coronavirus (MERS-CoV) Spike S1 Subunit Protein on the Phenotype of Camel Monocyte-Derived Macrophages. BIOLOGY 2025; 14:292. [PMID: 40136548 PMCID: PMC11940123 DOI: 10.3390/biology14030292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is an emerging zoonotic pathogen with different pathogenesis in humans and camels. The mechanisms behind the higher tolerance of camels to MERS-CoV infection are still unknown. Monocytes are innate myeloid cells that are able, depending on the local stimulation in their microenvironment, to differentiate into different functional subtypes of macrophages with an impact on the adaptive immune response. Several in vitro protocols have been used to induce the differentiation of monocyte-derived macrophages (MDMs) in human and several veterinary species. Such protocols are not available for camel species. In the present study, monocytes were separated from camel blood and differentiated in vitro in the presence of different stimuli into MDM. Camel MDMs generated in the presence of a combined stimulation of monocytes with LPS and GM-CSF resulted in the development of an M1 macrophages phenotype with increased abundance of the antigen-presentation receptor MHCII molecules and a decreased expression of the scavenger receptor CD163. The expression pattern of the cell markers CD163, CD14, CD172a, CD44, and CD9 on MDM generated in the presence of the MERS-CoV S1 protein revealed similarity with M-CSF-induced MDM, suggesting the potential of the MERS-CoV S1 protein to induce an M2 macrophages phenotype. Similarly to the effect of M-CSF, MERS-CoV-S protein-induced MDMs showed enhanced phagocytosis activity compared to non-polarized or LPS/GM-CSF-polarized MDMs. Collectively, our study represents the first report on the in vitro generation of monocyte-derived macrophages (MDMs) in camels and the characterization of some phenotypic and functional properties of camel MDM under the effect of M1 and M2 polarizing stimuli. In addition, the results suggest a polarizing effect of the MERS-CoV S1 protein on camel MDMs, developing an M2-like phenotype with enhanced phagocytosis activity. To understand the clinical relevance of these in vitro findings on disease pathogenesis and camel immune response toward MERS-CoV infection, further studies are required.
Collapse
Affiliation(s)
- Jamal Hussen
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (A.I.A.A.-M.); (K.B.)
| | - Abdullah I. A. Al-Mubarak
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (A.I.A.A.-M.); (K.B.)
| | - Turke Shawaf
- Department of Clinical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
| | - Khulud Bukhari
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (A.I.A.A.-M.); (K.B.)
| | - Khaled R. Alkharsah
- Department of Microbiology, College of Medicine, Imam Abdulrahman Bin Faisal University (IAU), Dammam 34212, Saudi Arabia;
| |
Collapse
|
5
|
Meng T, Zhang Y, Wang H, Wu W, Peng W, Yue J, Huang C, Liu W, Liang C, Yang C, Chen J. Irf7 aggravates prostatitis by promoting Hif-1α-mediated glycolysis to facilitate M1 polarization. Cell Mol Life Sci 2025; 82:90. [PMID: 39985573 PMCID: PMC11846824 DOI: 10.1007/s00018-025-05608-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is a common disorder associated with voiding symptoms and pain in the pelvic or perineal area. Macrophages, particularly the pro-inflammatory M1 subtype, are crucial initiating of CP/CPPS. Interferon regulatory factor 7 (Irf7) has been implicated in promoting M1 polarization, contributing to the onset and progression of autoimmunity. However, the role of Irf7 in the etiology and progression of CP/CPPS remains unclear. METHOD We established the experimental autoimmune prostatitis (EAP) mouse model by subcutaneous injection of prostate antigen combined with complete Freund's adjuvant. Six weeks after the first immunization, we analyzed the prostates, spleen, and blood to assess the degree of prostate inflammation, Irf7 expression levels, glycolysis, and M1 polarization to evaluate whether Irf7 could exacerbate the development of EAP by enhancing Hif-1α transcription, thereby increasing glycolysis and M1 polarization. Further investigations included sh-Irf7 intervention, Dimethyloxalylglycine (a Hif-1α agonist), and in vitro M1 polarization experiments. We also employed ChIP assays, dual-luciferase reporter assays, and q-PCR to explore if Irf7 could directly interact with the Hif-1α promoter in macrophages. RESULTS In the EAP mouse and cell models, elevated Irf7 expression was observed in inflamed tissues and cells. Reducing Irf7 expression decreased M1 cell glycolysis by inhibiting the nuclear translocation of Hif-1α, thus mitigating M1 cell polarization. Additionally, Irf7 was identified as a transcription factor that regulates Hif-1α transcription by interacting with its promoter in macrophages, confirmed through ChIP and dual-luciferase assays. Co-culturing macrophage cells with 3T3 fibroblasts with reduced Irf7 levels resulted in decreased fibrosis, and a significant reduction in prostate tissue fibrosis was noted in mice with Irf7 knockdown. CONCLUSION Our findings indicate that Irf7 can contribute to the development and progression of CP/CPPS by promoting glycolysis, which can enhance both M1 polarization as well as interstitial fibrosis in the prostate. This process was found to be mediated by the upregulation of Hif-1α transcription, presenting new potential therapeutic targets for managing CP/CPPS.
Collapse
Affiliation(s)
- Tong Meng
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Yi Zhang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Huihui Wang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Weikang Wu
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Wei Peng
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Jiabin Yue
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Cong Huang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Wanqing Liu
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Chaozhao Liang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China.
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| | - Cheng Yang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China.
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| | - Jing Chen
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China.
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| |
Collapse
|
6
|
Dousdampanis P, Aggeletopoulou I, Mouzaki A. The role of M1/M2 macrophage polarization in the pathogenesis of obesity-related kidney disease and related pathologies. Front Immunol 2025; 15:1534823. [PMID: 39867890 PMCID: PMC11758166 DOI: 10.3389/fimmu.2024.1534823] [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: 11/27/2024] [Accepted: 12/19/2024] [Indexed: 01/28/2025] Open
Abstract
Obesity is a rapidly growing health problem worldwide, affecting both adults and children and increasing the risk of chronic diseases such as type 2 diabetes, hypertension and cardiovascular disease (CVD). In addition, obesity is closely linked to chronic kidney disease (CKD) by either exacerbating diabetic complications or directly causing kidney damage. Obesity-related CKD is characterized by proteinuria, lipid accumulation, fibrosis and glomerulosclerosis, which can gradually impair kidney function. Among the immune cells of the innate and adaptive immune response involved in the pathogenesis of obesity-related diseases, macrophages play a crucial role in the inflammation associated with CKD. In obese individuals, macrophages enter a pro-inflammatory state known as M1 polarization, which contributes to chronic inflammation. This polarization promotes tissue damage, inflammation and fibrosis, leading to progressive loss of kidney function. In addition, macrophage-induced oxidative stress is a key feature of CKD as it also promotes cell damage and inflammation. Macrophages also contribute to insulin resistance in type 2 diabetes by releasing inflammatory molecules that impair glucose metabolism, complicating the management of diabetes in obese patients. Hypertension and atherosclerosis, which are often associated with obesity, also contribute to the progression of CKD via immune and inflammatory pathways. Macrophages influence blood pressure regulation and contribute to vascular inflammation, particularly via the renin-angiotensin system. In atherosclerosis, macrophages accumulate in arterial plaques, leading to chronic inflammation and plaque instability, which may increase the risk of CVD in CKD patients. This review focuses on the involvement of macrophages in CKD and highlights their role as a critical link between CKD and other pathologies. Targeting macrophage polarization and the ensuing macrophage-induced inflammation could be an effective therapeutic strategy for CKD and related diseases and improve outcomes for patients with obesity-related kidney disease.
Collapse
Affiliation(s)
| | - Ioanna Aggeletopoulou
- Laboratory of Immunohematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Athanasia Mouzaki
- Laboratory of Immunohematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| |
Collapse
|
7
|
Han SC, Kang JI, Choi YK, Yang DAH, Kim KJ, Boo HJ, Yoon WJ, Kang HK, Yoo ES, Boo HJ. 3-Bromo-4,5-dihydroxybenzaldehyde Attenuates Allergic Contact Dermatitis by Generating CD4 +Foxp3 + T cells. In Vivo 2025; 39:201-209. [PMID: 39740923 PMCID: PMC11705120 DOI: 10.21873/invivo.13818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/30/2024] [Accepted: 10/14/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND/AIM Regulatory T cells (Tregs) play a crucial role in inflammatory responses by regulating the activity of various immune cells. M2 macrophages induced by IL-10 and TGF-β exhibit anti-inflammatory functions and induce Treg differentiation. Although the beneficial effects of 3-bromo-4,5-dihydroxybenzaldehyde (BDB) on various diseases have been widely reported, the mechanisms, through which it alleviates allergic contact dermatitis (ACD) via Tregs and macrophages, are not well understood. Therefore, this study aimed to explore whether BDB suppresses ACD and induces Treg generation. MATERIALS AND METHODS Mice were sensitized with 1% dinitrochlorobenzene (DNCB), followed by the application of 0.3% DNCB to their ears every 3 days for 31 days. BDB (100 mg/kg) was administered orally once daily throughout the 31 days. Cytokine and transcription factor expression were analyzed via real-time PCR and western blotting, while CD4+Foxp3+ T cell differentiation and T cell proliferation were evaluated using flow cytometry. RESULTS BDB exhibited therapeutic efficacy in mice with ACD. In this study, the administration of BDB promoted the upregulation of transforming growth factor beta (TGF-β)-dependent CD4+Foxp3+ T cells. BDB elicited T cell hypo-responsiveness and suppressed the expression of cytokines related to the Th1, Th2, and Th17 cell subsets. BDB-M2 macrophages directly mediated the differentiation of CD4+Foxp3+ T cells from CD4+ T cells and concurrently suppressed the proliferation of CD4+ T cells. CONCLUSION BDB augments M2 macrophage function and induction of Tregs confers effective protection against ACD in mice. Consequently, BDB may represent a promising therapeutic approach for the treatment of inflammatory skin diseases.
Collapse
Affiliation(s)
- Sang-Chul Han
- Department of Medicine, College of Medicine, Jeju National University, Jeju, Republic of Korea
| | - Jung-Il Kang
- Department of Medicine, College of Medicine, Jeju National University, Jeju, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - Youn Kyung Choi
- Department of Medicine, College of Medicine, Jeju National University, Jeju, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - DA Hee Yang
- Department of Medicine, College of Medicine, Jeju National University, Jeju, Republic of Korea
| | - Ki Ju Kim
- Yong-am-hae-su Center, Jeju Technopark, Jeju, Republic of Korea
| | - Ha Jeong Boo
- Yong-am-hae-su Center, Jeju Technopark, Jeju, Republic of Korea
| | - Weon-Jong Yoon
- Jeju Biodiversity Research Institute, Jeju Technopark, Jeju, Republic of Korea
| | - Hee-Kyoung Kang
- Department of Medicine, College of Medicine, Jeju National University, Jeju, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - Eun-Sook Yoo
- Department of Medicine, College of Medicine, Jeju National University, Jeju, Republic of Korea
| | - Hye-Jin Boo
- Department of Medicine, College of Medicine, Jeju National University, Jeju, Republic of Korea;
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
8
|
He Y, Zhang Y, Zhu S, Liu YF, Liu S, Xu YJ. Monomethyl Branched-Chain Fatty Acids Suppress M1 Macrophage Polarization via FABP4/PPAR-γ Signaling Pathway. Mol Nutr Food Res 2024; 68:e2400310. [PMID: 39318069 DOI: 10.1002/mnfr.202400310] [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: 04/28/2024] [Revised: 08/27/2024] [Indexed: 09/26/2024]
Abstract
SCOPE Monomethyl-branched chain fatty acids (mmBCFAs) are found in a variety of food sources and are of great interest due to their potent antiinflammatory properties. However, most of the current researches have concentrated on the relationship between mmBCFAs and intestinal inflammation, and there is a large gap in the biological mechanisms involved behind their antiinflammatory effects. METHODS AND RESULTS The present study examines the role of mmBCFAs in modulating macrophage polarization. The results demonstrate that iso-C16:0 significantly inhibits macrophages M1 proinflammatory polarization through regulating FABP4/PPAR-γ pathway. Proteomics and molecular biology experiments verify that metabolic reprogramming is involved in the inhibition of M1 macrophage, referring to the upregulation of fatty acid oxidation, TCA cycle, and oxidative phosphorylation, as well as downregulation of glycolytic flux. CONCLUSION In summary, this study offers a novel perspective on the antiinflammatory effects mediated by mmBCFAs.
Collapse
Affiliation(s)
- Yuan He
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Reacher Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No. 1800, Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China
| | - Yu Zhang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Reacher Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No. 1800, Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China
| | - Shuang Zhu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Reacher Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No. 1800, Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China
| | - Yuan-Fa Liu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Reacher Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No. 1800, Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China
| | - Sha Liu
- Wuxi Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, 214071, China
| | - Yong-Jiang Xu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Reacher Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, No. 1800, Lihu Road, Wuxi, Jiangsu, 214122, People's Republic of China
| |
Collapse
|
9
|
Palomino-Cano C, Moreno E, Irache JM, Espuelas S. Targeting and activation of macrophages in leishmaniasis. A focus on iron oxide nanoparticles. Front Immunol 2024; 15:1437430. [PMID: 39211053 PMCID: PMC11357945 DOI: 10.3389/fimmu.2024.1437430] [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: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Macrophages play a pivotal role as host cells for Leishmania parasites, displaying a notable functional adaptability ranging from the proinflammatory, leishmanicidal M1 phenotype to the anti-inflammatory, parasite-permissive M2 phenotype. While macrophages can potentially eradicate amastigotes through appropriate activation, Leishmania employs diverse strategies to thwart this activation and redirect macrophages toward an M2 phenotype, facilitating its survival and replication. Additionally, a competition for iron between the two entities exits, as iron is vital for both and is also implicated in macrophage defensive oxidative mechanisms and modulation of their phenotype. This review explores the intricate interplay between macrophages, Leishmania, and iron. We focus the attention on the potential of iron oxide nanoparticles (IONPs) as a sort of immunotherapy to treat some leishmaniasis forms by reprogramming Leishmania-permissive M2 macrophages into antimicrobial M1 macrophages. Through the specific targeting of iron in macrophages, the use of IONPs emerges as a promising strategy to finely tune the parasite-host interaction, endowing macrophages with an augmented antimicrobial arsenal capable of efficiently eliminating these intrusive microbes.
Collapse
Affiliation(s)
- Carmen Palomino-Cano
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Esther Moreno
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Juan M. Irache
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Medical Research Institute (IdiSNA), Pamplona, Spain
| | - Socorro Espuelas
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Medical Research Institute (IdiSNA), Pamplona, Spain
| |
Collapse
|
10
|
Qian Z, Xiong W, Mao X, Li J. Macrophage Perspectives in Liver Diseases: Programmed Death, Related Biomarkers, and Targeted Therapy. Biomolecules 2024; 14:700. [PMID: 38927103 PMCID: PMC11202214 DOI: 10.3390/biom14060700] [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: 04/28/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Macrophages, as important immune cells of the organism, are involved in maintaining intrahepatic microenvironmental homeostasis and can undergo rapid phenotypic changes in the injured or recovering liver. In recent years, the crucial role of macrophage-programmed cell death in the development and regression of liver diseases has become a research hotspot. Moreover, macrophage-targeted therapeutic strategies are emerging in both preclinical and clinical studies. Given the macrophages' vital role in complex organismal environments, there is tremendous academic interest in developing novel therapeutic strategies that target these cells. This review provides an overview of the characteristics and interactions between macrophage polarization, programmed cell death, related biomarkers, and macrophage-targeted therapies. It aims to deepen the understanding of macrophage immunomodulation and molecular mechanisms and to provide a basis for the treatment of macrophage-associated liver diseases.
Collapse
Affiliation(s)
- Zibing Qian
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
| | - Wanyuan Xiong
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
| | - Xiaorong Mao
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
- Department of Infectious Disease, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Junfeng Li
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
- Institute of Infectious Diseases, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Department of Hepatology, The First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
11
|
Sun H, Yang Y, Jin Y, Chen H, Li A, Chen X, Yin J, Cai J, Zhang L, Feng X, Wang Y, Xiong W, Tang C, Wan B. Novel nanocomposites improve functional recovery of spinal cord injury by regulating NF-κB mediated microglia polarization. CHEMICAL ENGINEERING JOURNAL 2024; 487:150633. [DOI: 10.1016/j.cej.2024.150633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
12
|
Lin C, Chu Y, Zheng Y, Gu S, Hu Y, He J, Shen Z. Macrophages: plastic participants in the diagnosis and treatment of head and neck squamous cell carcinoma. Front Immunol 2024; 15:1337129. [PMID: 38650924 PMCID: PMC11033442 DOI: 10.3389/fimmu.2024.1337129] [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: 11/12/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) rank among the most prevalent types of head and neck cancer globally. Unfortunately, a significant number of patients receive their diagnoses at advanced stages, limiting the effectiveness of available treatments. The tumor microenvironment (TME) is a pivotal player in HNSCC development, with macrophages holding a central role. Macrophages demonstrate diverse functions within the TME, both inhibiting and facilitating cancer progression. M1 macrophages are characterized by their phagocytic and immune activities, while M2 macrophages tend to promote inflammation and immunosuppression. Striking a balance between these different polarization states is essential for maintaining overall health, yet in the context of tumors, M2 macrophages typically prevail. Recent efforts have been directed at controlling the polarization states of macrophages, paving the way for novel approaches to cancer treatment. Various drugs and immunotherapies, including innovative treatments based on macrophages like engineering macrophages and CAR-M cell therapy, have been developed. This article provides an overview of the roles played by macrophages in HNSCC, explores potential therapeutic targets and strategies, and presents fresh perspectives on the future of HNSCC treatment.
Collapse
Affiliation(s)
- Chen Lin
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| | - Yidian Chu
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| | - Ye Zheng
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| | - Shanshan Gu
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Yanghao Hu
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| | - Jiali He
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| | - Zhisen Shen
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| |
Collapse
|
13
|
Hu Y, Tang L, Wang Z, Yan H, Yi X, Wang H, Ma L, Yang C, Ran J, Yu A. Inducing in situ M2 macrophage polarization to promote the repair of bone defects via scaffold-mediated sustained delivery of luteolin. J Control Release 2024; 365:889-904. [PMID: 37952829 DOI: 10.1016/j.jconrel.2023.11.015] [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: 07/14/2023] [Revised: 10/26/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023]
Abstract
Immunoregulation mediated bone tissue engineering (BTE) has demonstrated huge potential in promoting repair of critical-size bone defects (CSBDs). The trade-off between stable immunoregulation function and extended immunoregulation period has posed a great challenge to this strategy. Here, we reported a 3D porous biodegradable Poly(HEMA-co-3APBA)/LUT scaffold, in which reversible boronic acid ester bond was formed between the 3APBA moiety and the catechol moiety of luteolin (LUT). The boronic acid ester bond not only protected the bioactivity of LUT but also extended the release period of LUT. The rationale behind the phenomenon of sustained LUT release was explained using a classical transition state theory. In vitro/in vivo assays proved the immunoregulation function of the scaffold in inducing M2 polarization of both M0 and M1 Mφ. The crosstalk between the scaffold treated Raw 264.7 and BMSCs were also investigated through the in vitro co-culture assay. The results demonstrated that the scaffold could induce immunoregulation mediated osteogenic differentiation of BMSCs. In addition, CSBDs model of SD rats was also applied, and the corresponding data proved that the scaffold could accelerate new bone formation, therefore promoting repair of CSBDs. The as-prepared scaffold might be a promising candidate for repair of CSBDs in the future.
Collapse
Affiliation(s)
- Yan Hu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China
| | - Lixi Tang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Zheng Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China
| | - Honghan Yan
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Xinzeyu Yi
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China
| | - Huimin Wang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Liya Ma
- Core Facility of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Changying Yang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Jiabing Ran
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China; College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430000, China.
| |
Collapse
|
14
|
Pang L, Ding Z, Chai H, Shuang W. The causal relationship between immune cells and different kidney diseases: A Mendelian randomization study. Open Med (Wars) 2023; 18:20230877. [PMID: 38152332 PMCID: PMC10751893 DOI: 10.1515/med-2023-0877] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 12/29/2023] Open
Abstract
Studies have suggested that the progress of most kidney diseases from occurrence to course and subsequent related complications are closely related to inflammatory reaction. Increased common leukocytes count in the family (neutrophils, eosinophils, basophils, lymphocytes, etc.) are also involved in the tissue damage of kidney diseases. However, these studies are only traditional observational studies, which cannot prove whether there is a causal relationship between these four kinds of leukocytes count and kidney diseases. We aim to explore the causal relationship between these four kinds of leukocytes count and kidney diseases by Mendelian randomization (MR). Large sample size of the genome-wide association database of four cell traits (neutrophil, basophil, lymphocyte, and eosinophil cell counts) in the leukocyte family were used as exposure variables. The outcome variables were various renal diseases (including chronic renal failure, acute renal failure, hypertensive heart or/and kidney disease, hypertensive renal disease, disorders resulting from impaired renal tubular function, and type 1 diabetes with renal complications). The covariates used in multivariable MR are also four cell traits related to blood cells (neutrophil, basophil, lymphocyte, and eosinophil cell counts). Instrumental variables and single nucleotide polymorphic loci were identified (P < 5 × 10-8. Linkage disequilibrium R2 < 0.001). The causal relationships were studied by inverse variance weighted (IVW), weighted median, and MR-Egger regression. Sensitivity analysis was also performed. In our study, IVW analysis results showed that increased neutrophil cell count was a risk factor for chronic renal failure (OR = 2.0245861, 95% CI = 1.1231207-3.649606, P = 0.01896524), increased basophil cell count was a risk factor for chronic renal failure (OR = 3.975935, 95% CI = 1.4871198-10.62998, P = 0.005942755). Basophil cell count was not a risk factor for acute renal failure (OR = 1.160434, 95% CI = 0.9455132-1.424207, P = 0.15448828). Increased basophil cell count was a protective factor for hypertensive heart and/or renal disease (OR = 0.7716065, 95% CI = 0.6484979-0.9180856, P = 0.003458707). Increased basophil cell count was a risk factor for disorders resulting from impaired renal tubular function (OR = 1.648131, 95% CI = 1.010116-2.689133, P = 0.04546835). Increased lymphocyte cell count was a risk factor for hypertensive renal disease (OR = 1.372961, 95% CI = 1.0189772-1.849915, P = 0.03719874). Increased eosinophil cell count was a risk factor for type 1 diabetes with renal complications (OR = 1.516454, 95% CI = 1.1826453-1.944482, P = 0.001028964). Macrophage inflammatory protein 1b levels was a protective factor for renal failure (OR = 0.9381862, 95% CI = 0.8860402-0.9934013, P = 0.02874872). After multivariable MR was used to correct covariates (neutrophil, basophil, and lymphocyte cell counts), the correlation effect between increased eosinophil cell counts and type 1 diabetes with renal complications was still statistically significant (P = 0.02201152). After adjusting covariates (neutrophil, basophil, and eosinophil cell counts) with multivariable MR, the correlation effect between increased lymphocyte cell counts and hypertensive renal disease was still statistically significant (P = 0.02050226). This study shows that increased basophils can increase the relative risk of chronic renal failure and renal tubular dysfunction, and reduce the risk of hypertensive heart disease and/or hypertensive nephropathy, while increased basophil cell count will not increase the relative risk of acute renal failure, increased neutrophil cell count can increase the risk of chronic renal failure, increased lymphocyte cell count can increase the relative risk of hypertensive nephropathy, and increased eosinophil cell count can increase the relative risk of type 1 diabetes with renal complications. Macrophage inflammatory protein 1b levels was a protective factor for renal failure.
Collapse
Affiliation(s)
- Lei Pang
- Department of Urology, The Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan City, 030012, Shanxi Province, China
- The First Clinical Medical College of Shanxi Medical University, Taiyuan City, 030012, Shanxi Province, China
| | - Zijun Ding
- Department of Neonatology, Shanxi Children's Hospital, Taiyuan City, 030013, Shanxi Province, China
| | - Hongqiang Chai
- Department of Urology, The Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan City, 030012, Shanxi Province, China
| | - Weibing Shuang
- The First Clinical Medical College of Shanxi Medical University, No. 85, Jiefang South Road, Yingze District, Taiyuan City, 030012, Shanxi Province, China
- Department of Urology, The First Hospital of Shanxi Medical University, No. 85, Jiefang South Road, Yingze District, Taiyuan City, 030012, Shanxi Province, China
| |
Collapse
|
15
|
Gu X, Shen H, Zhu G, Li X, Zhang Y, Zhang R, Su F, Wang Z. Prognostic Model and Tumor Immune Microenvironment Analysis of Complement-Related Genes in Gastric Cancer. J Inflamm Res 2023; 16:4697-4711. [PMID: 37872955 PMCID: PMC10590588 DOI: 10.2147/jir.s422903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023] Open
Abstract
Introduction The complement system is integral to the innate and adaptive immune response, helping antibodies eliminate pathogens. However, the potential role of complement and its modulators in the tumor microenvironment (TME) of gastric cancer (GC) remains unclear. Methods This study assessed the expression, frequency of somatic mutations, and copy number variations of complement family genes in GC derived from The Cancer Genome Atlas (TCGA). Lasso and Cox regression analyses were conducted to develop a prognostic model based on the complement genes family, with the training and validation sets taken from the TCGA-GC cohort (n=371) and the International Gene Expression Omnibus (GEO) cohort (n=433), correspondingly. The nomogram assessment model was used to predict patient outcomes. Additionally, the link between immune checkpoints, immune cells, and the prognostic model was investigated. Results In contrast to patients at low risk, those at high risk had a less favorable outcome. The prognostic model-derived risk score was shown to serve as a prognostic marker of GC independently, as per the multivariate Cox analysis. Nomogram assessment showed that the model had high reliability for predicting the survival of patients with GC in the 1, 3, 5 years. Additionally, the risk score was positively linked to the expression of immune checkpoints, notably CTLA4, LAG3, PDCD1, and CD274, according to an analysis of immune processes. The core gene C5aR1 in the prognostic model was found to be upregulated in GC tissues in contrast to adjoining normal tissues, and patients with elevated expressed levels of C5aR1 had lower 10-year overall survival (OS) rates. Conclusion Our work reveals that complement genes are associated with the diversity and complexity of TME. The complement prognosis model help improves our understanding of TME infiltration characteristics and makes immunotherapeutic strategies more effective.
Collapse
Affiliation(s)
- Xianhua Gu
- Department of Gynecology Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Honghong Shen
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Guangzheng Zhu
- Department of Surgical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Xinwei Li
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Yue Zhang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Rong Zhang
- Department of Gynecology Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Fang Su
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Zishu Wang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| |
Collapse
|
16
|
Medrano-Bosch M, Simón-Codina B, Jiménez W, Edelman ER, Melgar-Lesmes P. Monocyte-endothelial cell interactions in vascular and tissue remodeling. Front Immunol 2023; 14:1196033. [PMID: 37483594 PMCID: PMC10360188 DOI: 10.3389/fimmu.2023.1196033] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
Monocytes are circulating leukocytes of innate immunity derived from the bone marrow that interact with endothelial cells under physiological or pathophysiological conditions to orchestrate inflammation, angiogenesis, or tissue remodeling. Monocytes are attracted by chemokines and specific receptors to precise areas in vessels or tissues and transdifferentiate into macrophages with tissue damage or infection. Adherent monocytes and infiltrated monocyte-derived macrophages locally release a myriad of cytokines, vasoactive agents, matrix metalloproteinases, and growth factors to induce vascular and tissue remodeling or for propagation of inflammatory responses. Infiltrated macrophages cooperate with tissue-resident macrophages during all the phases of tissue injury, repair, and regeneration. Substances released by infiltrated and resident macrophages serve not only to coordinate vessel and tissue growth but cellular interactions as well by attracting more circulating monocytes (e.g. MCP-1) and stimulating nearby endothelial cells (e.g. TNF-α) to expose monocyte adhesion molecules. Prolonged tissue accumulation and activation of infiltrated monocytes may result in alterations in extracellular matrix turnover, tissue functions, and vascular leakage. In this review, we highlight the link between interactions of infiltrating monocytes and endothelial cells to regulate vascular and tissue remodeling with a special focus on how these interactions contribute to pathophysiological conditions such as cardiovascular and chronic liver diseases.
Collapse
Affiliation(s)
- Mireia Medrano-Bosch
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Blanca Simón-Codina
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Wladimiro Jiménez
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Elazer R. Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Pedro Melgar-Lesmes
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
17
|
Antar SA, Abd-Elsalam M, Abdo W, Abdeen A, Abdo M, Fericean L, Raslan NA, Ibrahim SF, Sharif AF, Elalfy A, Nasr HE, Zaid AB, Atia R, Atwa AM, Gebba MA, Alzokaky AA. Modulatory Role of Autophagy in Metformin Therapeutic Activity toward Doxorubicin-Induced Nephrotoxicity. TOXICS 2023; 11:273. [PMID: 36977038 PMCID: PMC10052439 DOI: 10.3390/toxics11030273] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 06/18/2023]
Abstract
Doxorubicin (DOX) is a frequent chemotherapeutic drug used to treat various malignant tumors. One of the key factors that diminish its therapeutic importance is DOX-induced nephrotoxicity. The first-line oral antidiabetic drug is metformin (Met), which also has antioxidant properties. The purpose of our study was to investigate the underlying molecular mechanisms for the potential protective effects of Met on DOX-triggered nephrotoxicity. Four animal groups were assigned as follows; animals received vehicle (control group), 200 mg/kg Met (Met group), DOX 15 mg/kg DOX (DOX group), and a combination of DOX and Met (DOX/Met group). Our results demonstrated that DOX administration caused marked histological alterations of widespread inflammation and tubular degeneration. Notably, the DOX-induced dramatic up-regulation of the nuclear factor-kappa B/P65 (NF-κB/P65), microtubule-associated protein light chain 3B (LC3B), neutrophil gelatinase-associated lipocalin (NGAL), interleukin-1beta (IL-1β), 8-hydroxy-2' -deoxyguanosine (8-OHdG), and Beclin-1 in renal tissue. A marked increase in the malondialdehyde (MDA) tissue level and a decrease in the total antioxidant capacity (TAC) were also recorded in DOX-exposed animals. Interestingly, Met could minimize all histopathological changes as well as the disruptions caused by DOX in the aforementioned measures. Thus, Met provided a workable method for suppressing the nephrotoxicity that occurred during the DOX regimen via the deactivation of the Beclin-1/LC3B pathway.
Collapse
Affiliation(s)
- Samar A. Antar
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24016, USA
| | - Marwa Abd-Elsalam
- Department of Histology, Faculty of Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Walied Abdo
- Department of Pathology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt
| | - Mohamed Abdo
- Department of Animal Histology and Anatomy, School of Veterinary Medicine, Badr University in Cairo (BUC), Badr City 32897, Egypt
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32897, Egypt
| | - Liana Fericean
- Department of Biology and Plant Protection, Faculty of Agriculture, University of Life Sciences “King Michael I” from Timișoara, Calea Aradului 119, CUI 3487181, 300645 Timisoara, Romania
| | - Nahed A. Raslan
- Clinical Pharmacy Program, College of Health Sciences and Nursing, Al-Rayan Colleges, Medina 42541, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11651, Egypt
| | - Samah F. Ibrahim
- Department of Clinical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Asmaa F. Sharif
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Tanta University, Tanta 31111, Egypt
| | - Amira Elalfy
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha 13518, Egypt
| | - Hend E. Nasr
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha 13518, Egypt
| | - Ahmed B. Zaid
- Department of Clinical Pathology, National Liver Institute, Menoufia University, Shibin Elkom 32511, Egypt
| | - Rania Atia
- Department of Physiology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
- Department of Basic Medical Science, Faculty of Applied Medical Science, Al-Baha University, Al-Baha 65779, Saudi Arabia
| | - Ahmed M. Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo 11829, Egypt
| | - Mohammed A. Gebba
- Department of Anatomy and Embryology, Faculty of Medicine, Benha University, Benha 13518, Egypt
| | - Amany A. Alzokaky
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11651, Egypt
| |
Collapse
|
18
|
Yang X, Liu X, Li J, Zhang P, Li H, Chen G, Zhang W, Wang T, Frazer I, Ni G. Caerin 1.1/1.9 Enhances Antitumour Immunity by Activating the IFN-α Response Signalling Pathway of Tumour Macrophages. Cancers (Basel) 2022; 14:cancers14235785. [PMID: 36497272 PMCID: PMC9738106 DOI: 10.3390/cancers14235785] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/09/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Macrophages are one of the essential components of the tumour microenvironment (TME) of many cancers and show complex heterogeneity and functions. More recent research has been focusing on the characterisation of tumour-associated macrophages (TAMs). Previously, our study demonstrated that caerin 1.1/1.9 peptides significantly improve the therapeutic efficacy of combined specific immunotherapy and immune checkpoint blockade in a murine transplantable tumour model (TC-1). In this study, the mice inoculated with TC-1 tumour were immunised differently. The TAMs were isolated using flow cytometry and characterised by cytokine ELISA. The survival rates of mice with different treatments containing caerin 1.1/19 were assessed comparatively, including those with/without macrophage depletion. The single-cell RNA sequencing (scRNA-seq) data of previous studies were integrated to further reveal the functions of TAMs with the treatments containing caerin 1.1/1.9. As a comparison, the TAMs of stage I and II cervical cancer patients were analysed using scRNA-seq analysis. We demonstrate that caerin induced tumour clearance is associated with infiltration of tumours by IL-12 secreting Ly6C+F4/80+ macrophages exhibiting enhanced IFN-α response signalling, renders animals resistant to further tumour challenge, which is lost after macrophage depletion. Our results indicate that caerin 1.1/1.9 treatment has great potential in improving current immunotherapy efficacy.
Collapse
Affiliation(s)
- Xiaodan Yang
- The First Affiliated Hospital, Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Xiaosong Liu
- The First Affiliated Hospital, Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan 528000, China
| | - Junjie Li
- The First Affiliated Hospital, Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Pingping Zhang
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan 528000, China
| | - Hejie Li
- School of Science, Engineering and Technology, University of the Sunshine Coast, Maroochydore BC, QLD 4558, Australia
| | - Guoqiang Chen
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan 528000, China
| | - Wei Zhang
- The First Affiliated Hospital, Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Tianfang Wang
- School of Science, Engineering and Technology, University of the Sunshine Coast, Maroochydore BC, QLD 4558, Australia
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD 4558, Australia
| | - Ian Frazer
- Faculty of Medicine, University of Queensland Diamantina Institute, Translational Research Institute, the University of Queensland, Woolloongabba, QLD 4102, Australia
- Correspondence: (I.F.); (G.N.)
| | - Guoying Ni
- The First Affiliated Hospital, Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou 510080, China
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan 528000, China
- Correspondence: (I.F.); (G.N.)
| |
Collapse
|
19
|
Hou Y, Shi J, Guo Y, Shi G. Inhibition of angiogenetic macrophages reduces disc degeneration-associated pain. Front Bioeng Biotechnol 2022; 10:962155. [PMID: 36304897 PMCID: PMC9592909 DOI: 10.3389/fbioe.2022.962155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/23/2022] [Indexed: 12/01/2022] Open
Abstract
Abnormal angiogenesis and innervation in avascular discs during lumbar disc degeneration (LDD) cause severe back pain. These pathological alterations in the degenerating discs are induced by cytokines partially produced and secreted by inflammatory cells, among which macrophages are the most frequently ones detected at the legion site. However, the role of macrophages as well as their polarization in regulation of innervation and angiogenesis in the degenerating discs is unclear. In this study, we analyzed macrophages in the degenerating discs from patients and detected a specific macrophage subtype that expresses high levels of vascular endothelial growth factor A (VEGF-A). Co-expression of M2 macrophage markers in this macrophage subtype suggested that they were a M2d-like subtype. High levels of VEGF-A and genes associated with angiogenesis were also detected in LDD specimens compared to control heathy discs from a public database, consistent with our finding. Moreover, the levels of VEGF-A in disc macrophages were strongly correlated to the pain score of the examined patients, but not to the Thompson classification of the degeneration level of the patients. In vitro, overexpressing VEGF-A in macrophages increased the tube formation, proliferation and migration of co-cultured endothelial cells, and increased the innervation of embryonic spinal cord explant into the co-cultured area for macrophages and skeletal myocytes. In vivo, an orthotopic injection of adeno-associated virus carrying siRNA for VEGF-A under a macrophage-specific CD68 promoter significantly reduced the number of VEGF-A-positive disc macrophages and alleviated the pain in LDD-mice. Together, these data suggest that inhibition of angiogenetic potential of macrophages may reduce disc degeneration-associated pain through suppression of angiogenesis and innervation, as a promising therapy for LDD-associated pain.
Collapse
|
20
|
Harmful and Beneficial Role of ROS 2020. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9873652. [PMID: 35910844 PMCID: PMC9337959 DOI: 10.1155/2022/9873652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022]
|
21
|
Wang S, Liu G, Li Y, Pan Y. Metabolic Reprogramming Induces Macrophage Polarization in the Tumor Microenvironment. Front Immunol 2022; 13:840029. [PMID: 35874739 PMCID: PMC9302576 DOI: 10.3389/fimmu.2022.840029] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/09/2022] [Indexed: 12/18/2022] Open
Abstract
Macrophages are one of the most important cells in the innate immune system, they are converted into two distinct subtypes with completely different molecular phenotypes and functional features under different stimuli of the microenvironment: M1 macrophages induced by IFN-γ/lipopolysaccharides(LPS) and M2 macrophages induced by IL-4/IL-10/IL-13. Tumor-associated macrophages (TAMs) differentiate from macrophages through various factors in the tumor microenvironment (TME). TAMs have the phenotype and function of M2 macrophages and are capable of secreting multiple cytokines to promote tumor progression. Both tumor cells and macrophages can meet the energy needs for rapid cell growth and proliferation through metabolic reprogramming, so a comprehensive understanding of pro-tumor and antitumor metabolic switches in TAM is essential to understanding immune escape mechanisms. This paper focuses on the functions of relevant signaling pathways and cytokines during macrophage polarization and metabolic reprogramming, and briefly discusses the effects of different microenvironments and macrophage pathogenicity, in addition to describing the research progress of inhibitory drugs for certain metabolic and polarized signaling pathways.
Collapse
Affiliation(s)
- Shilin Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Guohong Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yirong Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yunbao Pan
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
22
|
Wegher LSM, Kazmarek LM, Silva ACF, Maciel MG, Sasso BM, Teixeira F, Cintra ML. What is the Role of Different Macrophage Subsets in the Evolution of Juvenile Xanthogranulomas? Appl Immunohistochem Mol Morphol 2022; 30:e54-e58. [PMID: 35435864 DOI: 10.1097/pai.0000000000001029] [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: 01/12/2022] [Accepted: 02/26/2022] [Indexed: 11/26/2022]
Abstract
There are several activated forms of macrophages: 2 main groups are designated M1 and M2. While M1 macrophages have proinflammatory, bactericidal, and phagocytic functions and are the dominant phenotype observed in the early stages of inflammation, M2 macrophages are involved in constructive processes such as tissue repair; they play a role in wound healing and are required for revascularization and re-epithelialization. Juvenile xanthogranuloma (JXG) is the most common non-Langerhans cell histiocytosis. Its pathogenesis is not well understood, but it is not considered a neoplastic entity. JXGs possibly appear as a reaction to a nonspecific injury such as trauma or viral infection, although a genetic predisposition has been suggested in some cases. Tissue damage leads to a histiocytic response. JXGs appear, evolve toward maturation, and then most of them spontaneously regress. Young JXGs are characterized by small macrophages scattered in the dermis, in apposition close to the epidermis. As the lesion matures, the number of foamy macrophages and Touton cells increases and other cell types such as plasma cells, lymphocytes, and polymorphs are observed. Regressing xanthogranulomas will show numerous spindle cells in Significant values are in bold.a storiform distribution, interstitial fibrosis, and few foamy and Touton cells. In this study, we studied the immunophenotypic profile of macrophages found in cutaneous JXGs according to their stage of maturation. We examined the skin biopsies from 25 patients; all were embedded in paraffin and stained with hematoxylin and eosin and for immunohistochemistry. Typically, all JXGs were positive for factor XIIIa and CD4, and were negative for CD1a. The following histiocyte markers were used: CD68, CD204, CD163, MAC387, and HAM56. Images were analyzed by Image J software; data were statistically evaluated by SAS 9.0 software. The cases showed a slight predominance of males and the preference of the JXGs for the axial skin. Lesions occupied the papillary and reticular dermis in 85% of the cases and extended to the subcutaneous fat in the remainder. Compared with mature and regressing JXGs, younger lesions had a higher density of M1 macrophages, stained with MAC387. This antibody labels the histiocytes that have recently arrived in the areas of inflammation. As the lesions matured, there was an overwhelming predominance of M2 macrophages. These cells tended to cluster against the epidermis, except in the 2 cases in phase of regression. This suggests that there is a cross-talk between the epidermis and macrophages and that receptors, cytokines, chemokines, and adhesion molecules may play a role in the development and evolution of JXGs. These results indicate that, for most of their life, JXGs are formed by repairing M2 macrophages and are not just an M1 macrophagic response to a local antigen. The process appears to be influenced by chemical-mediator epidermal-macrophage cross-talking, considering the tendency of these cells to accumulate against the dermoepidermal junction.
Collapse
Affiliation(s)
- Lissa S M Wegher
- Departments of Pathology
- Dermatology, School of Medical Sciences, State University of Campinas, Sao Paulo, Brazil
| | | | | | - Marina G Maciel
- Dermatology, School of Medical Sciences, State University of Campinas, Sao Paulo, Brazil
| | - Bruna M Sasso
- Departments of Pathology
- Dermatology, School of Medical Sciences, State University of Campinas, Sao Paulo, Brazil
| | | | | |
Collapse
|
23
|
Meira CS, Soares JWC, Dos Reis BPZC, Pacheco LV, Santos IP, Silva DKC, de Lacerda JC, Daltro SRT, Guimarães ET, Soares MBP. Therapeutic Applications of Physalins: Powerful Natural Weapons. Front Pharmacol 2022; 13:864714. [PMID: 35450054 PMCID: PMC9016203 DOI: 10.3389/fphar.2022.864714] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/18/2022] [Indexed: 11/28/2022] Open
Abstract
Physalins, or 16,24-cyclo-13,14-seco steroids, are compounds belonging to the class of withanolides that can be found in plants of Solanaceae family, mainly in species belonging to the genus Physalis spp., which are annual herbaceous plants widely distributed in tropical and subtropical regions of the world. Physalins are versatile molecules that act in several cell signaling pathways and activate different mechanisms of cell death or immunomodulation. A number of studies have shown a variety of actions of these compounds, including anticancer, anti-inflammatory, antiparasitic, antimicrobial, antinociceptive, and antiviral activities. Here we reviewed the main findings related to the anticancer, immunomodulatory, and antiparasitic activities of physalins and its mechanisms of action, highlighting the \challenges and future directions in the pharmacological application of physalins.
Collapse
Affiliation(s)
- Cássio Santana Meira
- SENAI Institute of Innovation in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador, Brazil.,Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Brazil.,Department of Life Sciences, State University of Bahia (UNEB), Salvador, Brazil
| | | | | | | | | | | | - Julia Costa de Lacerda
- Bahiana School of Medicine and Public Health, Bahiana Foundation for the Development of Sciences, Salvador, Brazil
| | | | - Elisalva Teixeira Guimarães
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Brazil.,Department of Life Sciences, State University of Bahia (UNEB), Salvador, Brazil
| | - Milena Botelho Pereira Soares
- SENAI Institute of Innovation in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador, Brazil.,Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Brazil
| |
Collapse
|
24
|
Effects of CB2 Receptor Modulation on Macrophage Polarization in Pediatric Celiac Disease. Biomedicines 2022; 10:biomedicines10040874. [PMID: 35453624 PMCID: PMC9029516 DOI: 10.3390/biomedicines10040874] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/04/2022] [Accepted: 04/07/2022] [Indexed: 01/27/2023] Open
Abstract
Celiac Disease (CD) represents an autoimmune disorder triggered by the exposure to gluten in genetically susceptible individuals. Recent studies suggest the involvement of macrophages in CD pathogenesis. Macrophages are immune cells, present as pro-inflammatory classically activated macrophages (M1) or as anti-inflammatory alternatively activated macrophages (M2). The Cannabinoid Receptor 2 (CB2) has important anti-inflammatory and immunoregulatory properties. We previously demonstrated that a common CB2 functional variant, Q63R, causing CB2 reduced function, is associated with several inflammatory and autoimmune diseases The first aim of this study was to investigate the phenotype of macrophages isolated from peripheral blood of CD patients and CB2 expression. The second aim was to evaluate the effects of CB2 pharmacological modulation on CD macrophage polarization. Moreover, by an in vitro model of “immunocompetent gut” we investigated the role of CD macrophages in inducing intestinal barrier damage and the possibility to restore its functionality modulating their polarization. We found an increased expression of M1 macrophages and a CB2 reduced expression. We also demonstrated CD M1 macrophages in inducing the typical mucosal barrier damage of CD. CB2 stimulation switches macrophage polarization towards the anti-inflammatory M2 phenotype thus reducing inflammation but also limiting the epithelial dysfunction. Therefore, we suggest CB2 receptor as a possible novel therapeutic target for CD by regulating macrophages polarization and by preventing mucosal barrier damage.
Collapse
|
25
|
Yang W, Su G, Liu Y. Silencing p62 reduces ox-LDL-induced M1 polarization and inflammation in macrophages by inhibiting mTOR/NF-κB signaling pathways. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221110348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Macrophages can change their phenotypes according to the changes in the microenvironment, and thus have various functions, that is, macrophages polarization. Macrophage phenotype is associated with the progression of atherosclerotic plaques. Studies have shown a large accumulation of p62 protein in atherosclerotic plaques. Whether the accumulation of p62 protein affects the level of macrophage polarization and inflammation and its mechanism is not clear. The p62 levels of macrophages treated with ox-LDL were detected by western blotting and qRT-PCR. Several polarizing markers and cytokines associated with atherosclerosis were detected by western blotting, ELISA, qRT-PCR, and flow cytometry to assess macrophage phenotype. The effect of p62 on the treatment of macrophage polarization by ox-LDL was studied by silencing p62 by gene silencing technique. The activity of mTOR and NF-κB signaling pathways was evaluated by detecting p-mTOR and intranuclear p65 levels in western blotting to explore the mechanism of p62. Rapamycin inhibited mTOR to demonstrate its role in activating the NF-κB signaling pathway and in ox-LDL therapy of p62 induced M1 polarization in macrophages. ox-LDL induced a significant increase in p62 and an increase in M1 markers and inflammatory cytokines. After p62 silencing, M1 markers and inflammatory cytokines decreased significantly, while M2 markers and anti-inflammatory cytokines increased significantly. Silencing p62 inhibited p-mTOR and p65 nuclear translocation. Rapamycin inhibited ox-LDL-induced p65 nuclear translocation and M1 markers, and increased M2 markers. p62 protein accumulation in ox-LDL treatment macrophages induces M1 polarization and inflammatory cytokines through the mTOR/NF-κB pathway.
Collapse
Affiliation(s)
- Wei Yang
- Department of Laboratory Diagnostics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Laboratory Diagnosis, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guangming Su
- Department of Laboratory Diagnostics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yanhong Liu
- Department of Laboratory Diagnosis, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|