1
|
Du Z, Wei W, Lu S, Wang H, Feng C, Li Y, Cui X, Zhe J, Sun K, Liu K, Fan Q, Sun D, Bao W. PCL-PEtOx-based Crystalline-core Micelles for the Targeted Delivery of Paclitaxel and Trabectedin in Ovarian Cancer Therapy. Acta Biomater 2025:S1742-7061(25)00303-4. [PMID: 40306394 DOI: 10.1016/j.actbio.2025.04.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
Ovarian cancer (OC), which primarily metastasizes through ascites, is both invasive and fatal. Despite its toxicity and drug resistance, the platinum-based chemotherapy Taxol®+Carboplatin has been the first-line standard treatment for decades. Trabectedin (TBD) is a recently developed, highly effective antitumor drug that is also capable of regulating tumor-associated macrophages (TAMs), however, its severe side-effects hinder further clinical application. Here, we developed safe and efficient pH-responsive crystalline-core micelles for the combined treatment of OCs, exploiting parallel delivery of paclitaxel (PTX) and TBD. PCL-PEtOx-COOH was selected as the optimal carrier to encapsulate PTX or TBD, which self-assemble into micelles with internal crystalline cores. The carboxyl group exposed on the surface of the micelles was utilized to react with the amines of Herceptin and hyaluronic acid cross-linked polymer (Herceptin-HA) to form PTX(Target). Similarly, TBD(Target) was formed by reaction with the CD206-targeted peptide mUNO. The low critical micelle concentrations of PTX(Target) and TBD(Target) stabilize the micelles in the bloodstream and normal tissues to prevent drug release. In an acidic microenvironment, the tertiary amide group on PEtOx chain of micelles ionizes, causing disassembly and pH-responsive release. Compared with Taxol®+Carboplatin, the combination therapy displayed dramatically improved safety and efficacy, as evidenced by the elimination of peritoneal tumor spheroids and reduced expression of NOX4, a gene that is overexpressed in most OC tissues. Furthermore, in human tissues, the ROS-response gene NOX4 is linked to the development of M2-type TAMs. Collectively, this study provides a safe and effective non-platinum-based chemotherapy for OC, offering an alternative to traditional Taxol®+Carboplatin. STATEMENT OF SIGNIFICANCE: (1) Significance: This work reports a new approach for ovarian cancer (OC) treatment. We utilized trabectedin (TBD) which a recently developed, highly effective antitumor drug that is also capable of regulating tumor associated macrophages (TAMs) combined with paclitaxel (PTX) to replace platinum-based chemotherapy Taxol®+Carboplatin (TC regimen). Compared to the clinical formulations, Yondelis® and Taxol®, pH-responsive PCL-PEtOx-based crystalline-core micelles were utilized for targeted independent delivery of TBD and PTX to TAMs and tumor cells, which maintained safe and efficient transport, overcoming the challenges posed by TAMs and carboplatin resistance. The system capabilities have also been confirmed in organoid and PDX models. (2) This is the first report demonstrating that this approach simultaneously overcomes the abdominal metastasis and carboplatin resistance of OC.
Collapse
Affiliation(s)
- Zixiu Du
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| | - Wei Wei
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Shuli Lu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Hao Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
| | - Chenxu Feng
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Yinuo Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
| | - Xinyi Cui
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jianan Zhe
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910 Hengshan Road, Shanghai, 200030, China
| | - Kuo Sun
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, 1st Minde Road, Nanchang, Jiangxi, 330006, China
| | - Kuai Liu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Qiong Fan
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910 Hengshan Road, Shanghai, 200030, China.
| | - Donglei Sun
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China; Shanghai Key Laboratory for Antibody-Drug Conjugates with Innovative Target, Shanghai, 200240, China.
| | - Wei Bao
- Department of Obstetrics and Gynecology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China.
| |
Collapse
|
2
|
Xia X, Chen W, Zhou T, Zhou F, Lu C, Yan Z, Zhao Q, Su Q. TEPP-46 inhibits glycolysis to promote M2 polarization of microglia after ischemic stroke. Int Immunopharmacol 2025; 149:114148. [PMID: 39904037 DOI: 10.1016/j.intimp.2025.114148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/12/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Following an ischemic stroke, neuroinflammation is triggered and is often typified by microglial activation. According to recent research, increased glycolysis metabolism frequently occurs when microglia become activated in an inflammatory response. In this study, we found that the PKM2 expression of microglia was gradually increased during the activation of microglia in ischemic stroke. TEPP-46, the activator of PKM2, enhanced the M2 polarization and promoted phagocytosis of microglia both in vivo and in vitro. Meanwhile, TEPP-46 administration ameliorated neuroinflammation and neuronal injuries and reduced the infarct volume of tMCAO mice. Mechanistically, we demonstrated that TEPP-46 suppressed the nuclear translocation of PKM2 and the interaction of PKM2 and HIF-1α, and inhibited glycolysis of microglia. According to our research, PKM2 modulation in microglia may be a viable therapeutic approach to lessen neuroinflammation following ischemic stroke, and TEPP-46 may be able to polarize microglia from an M1 to an M2 phenotype after ischemia/reperfusion damage.
Collapse
Affiliation(s)
- Xiaomei Xia
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Kangda College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang 222000 China; Department of Rehabilitation Medicine, Kangda College of Nanjing Medical University, Lianyungang 222000 China
| | - Wenli Chen
- Department of Rehabilitation Medicine, ZhongDa Hospital Southeast University, Nanjing 210009 China
| | - Ting Zhou
- Department of Rehabilitation Medicine, Kangda College of Nanjing Medical University, Lianyungang 222000 China
| | - Fang Zhou
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Kangda College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang 222000 China
| | - Can Lu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Kangda College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang 222000 China
| | - Zhenzhuang Yan
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Kangda College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang 222000 China
| | - Qin Zhao
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Kangda College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang 222000 China.
| | - Qinglun Su
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Kangda College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang 222000 China.
| |
Collapse
|
3
|
Yao Y, Luo Y, Liang X, Zhong L, Wang Y, Hong Z, Song C, Xu Z, Wang J, Zhang M. The role of oxidative stress-mediated fibro-adipogenic progenitor senescence in skeletal muscle regeneration and repair. Stem Cell Res Ther 2025; 16:104. [PMID: 40025535 PMCID: PMC11872320 DOI: 10.1186/s13287-025-04242-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/18/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Stem cells play a pivotal role in tissue regeneration and repair. Skeletal muscle comprises two main stem cells: muscle stem cells (MuSCs) and fibro-adipogenic progenitors (FAPs). FAPs are essential for maintaining the regenerative milieu of muscle tissue and modulating the activation of muscle satellite cells. However, during acute skeletal muscle injury, the alterations and mechanisms of action of FAPs remain unclear. METHODS we employed the GEO database for bioinformatics analysis of skeletal muscle injury. A skeletal muscle injury model was established through cardiotoxin (CTX, 10µM, 50µL) injection into the tibialis anterior (TA) of C57BL/6 mice. Three days post-injury, we extracted the TA, isolated FAPs (CD31-CD45-PDGFRα+Sca-1+), and assessed the senescence phenotype through SA-β-Gal staining and Western blot. Additionally, we established a co-culture system to evaluate the capacity of FAPs to facilitate MuSCs differentiation. Finally, we alleviated the senescent of FAPs through in vitro (100 µM melatonin, 5 days) and in vivo (20 mg/kg/day melatonin, 15 days) administration experiments, confirming melatonin's pivotal role in the regeneration and repair processes of skeletal muscle. RESULTS In single-cell RNA sequencing analysis, we discovered the upregulation of senescence-related pathways in FAPs following injury. Immunofluorescence staining revealed the co-localization of FAPs and senescent markers in injured muscles. We established the CTX injury model and observed a reduction in the number of FAPs post-injury, accompanied by the manifestation of a senescent phenotype. Melatonin treatment was found to attenuate the injury-induced senescence of FAPs. Further co-culture experiments revealed that melatonin facilitated the restoration of FAPs' capacity to promote myoblast differentiation. Through GO and KEGG analysis, we found that the administration of melatonin led to the upregulation of AMPK pathway in FAPs, a pathway associated with antioxidant stress response. Finally, drug administration experiments corroborated that melatonin enhances skeletal muscle regeneration and repair by alleviating FAP senescence in vivo. CONCLUSION In this study, we first found FAPs underwent senescence and redox homeostasis imbalance after injury. Next, we utilized melatonin to enhance FAPs regenerative and repair capabilities by activating AMPK signaling pathway. Taken together, this work provides a novel theoretical foundation for treating skeletal muscle injury.
Collapse
Affiliation(s)
- Yuqing Yao
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Yusheng Luo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiaomei Liang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Li Zhong
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yannan Wang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-Sen University, Shenzhen, China
| | - Zhengchao Hong
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Chao Song
- School of Electronics and Communication Engineering, Shenzhen Campus of Sun Yat-sen University, Sun Yat-Sen University, Shenzhen, China
| | - Zeyu Xu
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-Sen University, Shenzhen, China
| | - Jiancheng Wang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Miao Zhang
- Department of Physical Education, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
4
|
Corken A, Weinkopff T, Wahl EC, Sikes JD, Thakali KM. Platelets Modulate Leukocyte Population Composition Within Perivascular Adipose Tissue. Int J Mol Sci 2025; 26:1625. [PMID: 40004089 PMCID: PMC11855773 DOI: 10.3390/ijms26041625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Perivascular adipose tissue (PVAT) regulates vascular tone and is composed of adipocytes and several leukocyte subpopulations. Diet can modify PVAT function, as obesogenic diets cause morphological changes to adipocytes and skew the leukocyte phenotype, leading to PVAT dysregulation and impaired vasoregulation. Of note, platelets, the clot-forming cells, also modulate many facets of leukocyte activity, such as tissue infiltration and polarity. We aimed to determine whether platelets regulate the leukocyte populations residing within PVAT. Male C57Bl/6J mice were fed a Western diet (30% kcal sucrose, 40% kcal fat, 8.0% sodium) to develop obesogenic conditions for PVAT leukocyte remodeling. Diet was either administered acutely (2 weeks) or extended (8 weeks) to gauge the length of challenge necessary for remodeling. Additionally, platelet depletion allowed for the assessment of platelet relevance in PVAT leukocyte remodeling. Abdominal PVAT (aPVAT) and thoracic PVAT (tPVAT) were then isolated and leukocyte composition evaluated by flow cytometry. Compared to control, Western diet alone did not significantly impact PVAT leukocyte composition for either diet length. Platelet depletion, independent of diet, significantly disrupted PVAT leukocyte content with monocytes/macrophages most impacted. Furthermore, tPVAT appeared more sensitive to platelet depletion than aPVAT, providing novel evidence of platelet regulation of leukocyte composition within PVAT depots.
Collapse
Affiliation(s)
- Adam Corken
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA;
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA; (E.C.W.); (J.D.S.)
| | - Tiffany Weinkopff
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Elizabeth C. Wahl
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA; (E.C.W.); (J.D.S.)
| | - James D. Sikes
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA; (E.C.W.); (J.D.S.)
| | - Keshari M. Thakali
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA;
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA; (E.C.W.); (J.D.S.)
| |
Collapse
|
5
|
Fennel ZJ, O'Connell RM, Drummond MJ. Macrophage immunometabolism: emerging targets for regrowth in aging muscle. Am J Physiol Endocrinol Metab 2025; 328:E186-E197. [PMID: 39763086 PMCID: PMC12079615 DOI: 10.1152/ajpendo.00403.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/31/2025]
Abstract
The recovery from muscle atrophy is impaired with aging as characterized by improper muscle remodeling and sustained functional deficits. Age-related deficits in muscle regrowth are tightly linked with the loss of early pro-inflammatory macrophage responses and subsequent cellular dysregulation within the skeletal muscle niche. Macrophage inflammatory phenotype is regulated at the metabolic level, highlighting immunometabolism as an emerging strategy to enhance macrophage responses and restore functional muscle regrowth. Accordingly, metabolic targets with an emphasis on glycolytic, hypoxia, and redox-related pathways stand out for their role in promoting macrophage inflammation and enhancing muscle regrowth in aging. Here we highlight promising immuno-metabolic targets that could be leveraged to restore optimal pro-inflammatory macrophage function in aging and enhance muscle regrowth following muscular atrophy.
Collapse
Affiliation(s)
- Zachary J Fennel
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, United States
| | - Ryan M O'Connell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, United States
| | - Micah J Drummond
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, United States
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
6
|
Phuong NQ, Bilal M, Nawaz A, Anh LD, Memoona, Aslam MR, Khalid S, Kado T, Watanabe Y, Nishimura A, Igarashi Y, Okabe K, Hirabayashi K, Yamamoto S, Nakagawa T, Mori H, Usui I, Fujisaka S, Hayashi R, Tobe K. Role of transforming growth factor-β1 in regulating adipocyte progenitors. Sci Rep 2025; 15:941. [PMID: 39824986 PMCID: PMC11748614 DOI: 10.1038/s41598-024-81917-7] [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] [Accepted: 11/29/2024] [Indexed: 01/20/2025] Open
Abstract
Adipose tissue (AT) metabolism involves coordinating various cells and cellular processes to regulate energy storage, release, and overall metabolic homeostasis. Therein, macrophage and its cytokine are important in controlling tissue homeostasis. Among cytokines, the role of transforming growth factor-β1 (Tgf-β1), a cytokine abundantly expressed in CD206+ M2-like macrophage and correlated with the expansion of AT and fibrosis, in AT metabolism, remains unknown. We used CD206CreERT2; Tgf-β1f/f mouse model in which the Tgf-β1 gene was conditionally deleted in CD206+ M2-like macrophages followed by tamoxifen administration, to investigate the role of the Tgf-β1 gene in glucose and insulin metabolism. Our data demonstrated that lack of CD206+ M2-like macrophages derived Tgf-β1 gene improved glucose metabolism and insulin sensitivity by enhancing adipogenesis via hyperplasia. The Tgf-β1 gene, specifically from CD206+ M2-like macrophages, deletion stimulated APs' proliferation and differentiation, leading to the generation of smaller mature adipocytes, therefore enhancing insulin sensitivity and improving glucose metabolism under normal chow conditions. Our study brings a new perspective that Tgf-β1 gene deletion specific from CD206+ M2-like macrophage promotes adipocyte hyperplasia, improving glucose homeostasis and insulin sensitivity in the lean state.
Collapse
Affiliation(s)
- Nguyen Quynh Phuong
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
- Clinical Oncology, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Muhammad Bilal
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
- Research Center for Pre-Disease Science, Faculty of Education and Research Promotion, University of Toyama, Toyama, 930-0194, Japan.
- Advanced Postdoctoral Fellowships of the Japan Diabetes Society (JDS), Tokyo, Japan.
| | - Allah Nawaz
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Le Duc Anh
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Memoona
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Muhammad Rahil Aslam
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Sana Khalid
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Tomonobu Kado
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Yoshiyuki Watanabe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Ayumi Nishimura
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
- Faculty of Education and Research Promotion, University of Toyama, Toyama, 930-0194, Japan
| | - Yoshiko Igarashi
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
- JSPS Research Fellowship for Young Scientist Japan, Tokyo, Japan
| | - Keisuke Okabe
- Toyama University Hospital, Center for Clinical and Translational Research, Toyama, Japan
| | - Kenichi Hirabayashi
- Department of Diagnostic Pathology, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Seiji Yamamoto
- Department of Pathology, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Isao Usui
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Tochigi, Japan
| | - Shiho Fujisaka
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Ryuji Hayashi
- Clinical Oncology, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Kazuyuki Tobe
- Research Center for Pre-Disease Science, Faculty of Education and Research Promotion, University of Toyama, Toyama, 930-0194, Japan.
- Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|
7
|
Hu H, Yin Y, Zhou H, Jiang B, Cai T, Wu S, Guo S. Umbilical cord mesenchymal stem cell-derived exosomal Follistatin inhibits fibrosis and promotes muscle regeneration in mice by influencing Smad2 and AKT signaling. Exp Cell Res 2025; 444:114396. [PMID: 39732451 DOI: 10.1016/j.yexcr.2024.114396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 12/22/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Promoting muscle regeneration through stem cell therapy has potential risks. We investigated the effect of umbilical cord mesenchymal stem cells (UMSCs) Exosomes (Exo) Follistatin on muscle regeneration. METHODS The Exo was derived from UMSCs cells and was utilized to affect the mice muscle injury model and C2C12 cells myotubes atrophy model. The Western blot, qRT-PCR and IF were utilized to determine the effects of Exo on the levels of Follistatin, MyHC, MyoD, Myostatin, MuRF1, MAFbx, α-SMA, Collagen I, Smad2, and AKT. In addition, HE and Masson staining were used to assess muscle tissue damage in mice. RESULTS The level of Follistatin in Exo was significantly higher than that in UMSCs. UMSCs-Exo increased the levels of Follistatin, MyHC, MyoD, and p-Smad2 and decreased the levels of Myostatin, MuRF1, MAFbx, α-SMA, Collagen I, p-AKT, and p-mTOR in mice or C2C12 cells. In addition, UMSCs-Exo decreased levels of inflammation and fibrosis in mice. However, UMSCs-Exo-si-Follistatin reversed the effect of UMSCs-Exo. Transfection of oe-Smad2 up-regulated the protein levels of Collagen I, α-SMA, and changed the ratio of p-Smad2/Smad2 expression to 0.33, and 0.34, 0.73. LY294002 decreased the levels of MyHC, MyoD, and the ratio of p-AKT/AKT and p-mTOR/mTOR expression to 0.12, 0.17, 0.33, and 0.41, increased the levels of MuRF1 and MAFbx to 0.36 and 0.34. CONCLUSION This study demonstrated that Follistatin in UMSCs-Exo inhibits fibrosis and promotes muscle regeneration in mice by regulating Smad and AKT signaling.
Collapse
Affiliation(s)
- Hai Hu
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, China; Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, China
| | - Yuesong Yin
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hecheng Zhou
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Binbin Jiang
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ting Cai
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, China
| | - Song Wu
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, China.
| | - Shuangfei Guo
- Department of Limbs (Foot and Hand) Microsurgery, Chenzhou No.1 People's Hospital, The First Clinical Medical College Affiliated to Southern Medical University, Chenzhou, Hunan, China.
| |
Collapse
|
8
|
HANG W, WANG L, BO Y, ZUO S, WANG S, LI H, BU C, ZHAO J, ZHOU X. Bufei Huoxue capsule alleviates silicosis by inhibiting the activation of nucleotide-like receptor containing pyrin domain 3 inflammasome and macrophages polarization based on network pharmacology. J TRADIT CHIN MED 2024; 44:1236-1246. [PMID: 39617709 PMCID: PMC11589560 DOI: 10.19852/j.cnki.jtcm.20240626.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 03/08/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVE To predict the targets of Bufei Huoxue capsule (, BFHX) using network pharmacology analysis and to explore its effects and functional targets in a silicotic rat model. METHODS The drug and disease targets were correlated through network pharmacology analysis to explore the targets and signaling pathways of BFHX affecting silicosis. NR8383 cells were cultured to verify the core genes and pathways. A rat model of silicosis was established to verify whether the mechanism behind SiO2-caused pulmonary fibrosis was alleviated by BFHX (0.82 g/kg) and how it affected key targets and pathways. RESULTS Overlapping BFHX and silicotic gene targets produced 159 interactive targets, and 55 were screened by network topology analysis. The results of gene ontology and Kyoto encyclopedia of genes and genomes enrichment analyses suggested that BFHX could affect silicosis through the nucleotide-like receptor containing pyrin domain 3 (NLRP3) inflammasome. In NR8383 cells, the expression of core genes related to the NLRP3 inflammasome could be inhibited by BFHX treatment. BFHX reduced the degree of alveolitis and collagen deposition, attenuating pulmonary fibrosis in SiO2-induced rat model. Pulmonary macrophage pyroptosis after SiO2 exposure was observed under transmission electron microscopy. BFHX alleviated the morphological characteristics of pyroptosis. BFHX also reduced the expression of NLRP3, caspase-1, interleukin-1 beta (IL-1β), IL-18, IL-6, and tumor necrosis factor-alpha in lung tissues of silicotic rat model. BFHX affected the K ion content in bronchoalveolar lavage fluid when assessed by energy dispersive spectrometer testing. The expression of CD68+ and CD206+ were also reduced after BFHX intervention. CONCLUSION NOD-like receptor signaling is vital for BFHX's effects on silicosis. It exerts anti-pulmonary fibrosis effects by inhibiting pulmonary macrophage pyroptosis and polarization through NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Wenlu HANG
- 1 Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Lin WANG
- 1 Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Yun BO
- 2 Medical College of Anhui University of Science and Technology, Anhui 232001, China
| | - Shurun ZUO
- 1 Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Songquan WANG
- 3 School of Mechatronic Engineering, Jiangsu Normal University, Xuzhou 221000, China
| | - Haiquan LI
- 1 Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Chunlu BU
- 1 Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Jie ZHAO
- 1 Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Xianmei ZHOU
- 4 Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
9
|
Jiang C, Wang W, Chen YL, Chen JH, Zhang ZW, Li J, Yang ZC, Li XC. Macrophage polarization and macrophage-related factor expression in hypertrophy of the ligamentum flavum. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2024; 33:4476-4487. [PMID: 39375228 DOI: 10.1007/s00586-024-08513-1] [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: 01/30/2024] [Revised: 09/03/2024] [Accepted: 09/29/2024] [Indexed: 10/09/2024]
Abstract
PURPOSE Owing to the unknow types of infiltrating macrophages and the corresponded factors, we aimed to investigate the specific types of infiltrating macrophages involved in HLF and the expression of macrophage-related factors. METHODS The ligamentum flavum was obtained from patients with lumbar spinal stenosis (HLF group; n = 15) and lumbar disc herniation (non-hypertrophic ligamentum flavum [NLF] group; n = 15). Ligamentum flavum specimens were paraffin embedded, followed by histological and immunohistochemical staining to identify the macrophage type and expression of macrophage-related factors. RESULTS The HLF group demonstrated CD206 marker expression, while the NLF group did not (P < 0.0001; n = 11). CD68 marker was expressed in both groups (P > 0.05; n = 11). CCR7 was not expressed in either group. The expression levels of the extracellular matrix proteins aggrecan (Agg), type I collagen (Coll1), and type II collagen (Coll2) were higher in the HLF group than in the NLF group (P < 0.0001; n = 11). The aging markers p21, p16, and p53 were expressed in the HLF group, but not in the NLF group (P < 0.0001; n = 11). The expression levels of the inflammatory factors TNF-α and IL-1β were higher in the HLF group than in the NLF group (P < 0.0001; n = 11). Similarly, the expression level of the fibrosis factor TGF-β1 was higher in the HLF group than in the NLF group (P < 0.0001; n = 11). CONCLUSIONS The infiltration of M2 macrophages may be involved in HLF, while involvement of M1 macrophages may only occur early in inflammation. The expression of extracellular matrix proteins and macrophage-related factors was increased. Aging may also be associated with HLF.
Collapse
Affiliation(s)
- Cheng Jiang
- Department of Orthopedic Surgery, Gaozhou People's Hospital, No.89 XiGuan Rd, Gaozhou, 525200, Guangdong, China
- Central Laboratory of Orthopedics, Gaozhou People's Hospital, XiGuan Rd, Gaozhou, 525200, China
| | - Wei Wang
- Graduate School of Guangdong Medical University, No. 2, Wenming East Road, Zhanjiang, 524023, Guangdong, China
| | - Yong-Long Chen
- Graduate School of Guangdong Medical University, No. 2, Wenming East Road, Zhanjiang, 524023, Guangdong, China
| | - Jiong-Hui Chen
- Graduate School of Guangdong Medical University, No. 2, Wenming East Road, Zhanjiang, 524023, Guangdong, China
| | - Zhen-Wu Zhang
- Graduate School of Guangdong Medical University, No. 2, Wenming East Road, Zhanjiang, 524023, Guangdong, China
| | - Jun Li
- Graduate School of Guangdong Medical University, No. 2, Wenming East Road, Zhanjiang, 524023, Guangdong, China
| | - Zhi-Chao Yang
- Graduate School of Guangdong Medical University, No. 2, Wenming East Road, Zhanjiang, 524023, Guangdong, China
| | - Xiao-Chuan Li
- Department of Orthopedic Surgery, Gaozhou People's Hospital, No.89 XiGuan Rd, Gaozhou, 525200, Guangdong, China.
- Central Laboratory of Orthopedics, Gaozhou People's Hospital, XiGuan Rd, Gaozhou, 525200, China.
| |
Collapse
|
10
|
Jin J, Yang Y, Yang J, Sun Z, Wang D, Qin Y, Ruan C, Li D, Pan Y, Wu J, Zhang C, Hu Y, Lei P. Macrophage metabolic reprogramming-based diabetic infected bone defect/bone reconstruction though multi-function silk hydrogel with exosome release. Int J Biol Macromol 2024; 278:134830. [PMID: 39154694 DOI: 10.1016/j.ijbiomac.2024.134830] [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: 06/02/2024] [Revised: 07/15/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Diabetic infected bone defects (DIBD) with abnormal immune metabolism are prone to the hard-to-treat bacterial infections and delayed bone regeneration, which present significant challenges in clinic. Control of immune metabolism is believed to be important in regulating fundamental immunological processes. Here, we developed a macrophage metabolic reprogramming hydrogel composed of modified silk fibroin (Silk-6) and poly-l-lysine (ε-PL) and further integrated with M2 Macrophage-derived Exo (M2-Exo), named Silk-6/ε-PL@Exo. This degradable hydrogel showed a broad-spectrum antibacterial performance against both Gram-positive and -negative bacteria. More importantly, the release of M2-Exo from Silk-6/ε-PL@Exo could target M1 macrophages, modulating the activity of the key enzyme hexokinase II (HK2) to control the inflammation-related NF-κB pathway, alleviate lactate accumulation, and inhibit glycolysis to normalize the cycle, thereby promoting M1-to-M2 balance. Using a rat model of DIBD, Silk-6/ε-PL@Exo hydrogel promoted infection control, balanced immune responses and accelerated the bone defect healing. Overall, this study demonstrates that this Silk-6/ε-PL @Exo is a promising filler biomaterial with multi-function to treat DIBD and emphasizes the importance of metabolic reprogramming in bone regeneration.
Collapse
Affiliation(s)
- Jiale Jin
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yiqi Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jian Yang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zeyu Sun
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Dongyu Wang
- Department of Orthopedic Surgery, Xiangya Hospital Central South University, Changsha 410008, China
| | - Yifang Qin
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China
| | - Chengxin Ruan
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Dongdong Li
- Department of Orthopedic Surgery, Ningxia Medicial University, Yinchuan 200233, China
| | - Yi Pan
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiangdong Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Chi Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Yihe Hu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Pengfei Lei
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
11
|
Huang N, Zou K, Zhong Y, Luo Y, Wang M, Xiao L. Hotspots and trends in satellite cell research in muscle regeneration: A bibliometric visualization and analysis from 2010 to 2023. Heliyon 2024; 10:e37529. [PMID: 39309858 PMCID: PMC11415684 DOI: 10.1016/j.heliyon.2024.e37529] [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: 12/10/2023] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Background The incidence of muscle atrophy or sports injuries is increasing with time and population aging, thereby attracting considerable attention to muscle generation research. Muscle satellite cells, which play an important role in this process, lack comprehensive literature regarding their use for muscle regeneration. Hence, this study aimed to analyze the hotspots and trends in satellite cell research from 2010 to 2023, providing a reference for muscle regeneration research. Methods Studies on satellite cells' role in muscle regeneration from 2010 to 2023 were retrieved from the Web of Science Core Collection. Using CiteSpace and VOSviewer, we analyzed annual publications, authors and co-citing authors, countries and institutions, journals and co-citing journals, co-citing references, and keywords. Results From 2010 to 2023, 1468 papers were retrieved, indicating an overall increasing trend in the number of annual publications related to satellite cells in muscle regeneration. The United States had the highest number of publications, while the Institut National de la Santé et de la Recherche Médicale was the institution with the most publications. Among journals, " PloS One" had the highest number of published papers, and "Cell" emerged as the most co-cited journal. A total of 7425 authors were involved, with Michael A. Rudnicki being the author with the highest number of publications and the most co-cited author. The most cited reference was "Satellite cells and the muscle stem cell niche." Among keywords, "satellite cells" was the most common, with "heterogeneity" having the highest centrality. Frontier themes included "Duchenne muscular dystrophy," "skeletal muscle," "in-vivo," "muscle regeneration," "mice," "muscle atrophy," "muscle fibers," "inflammation," " mesenchymal stem cells," and "satellite cell." Conclusion This study presents the current status and trends in satellite cell research on muscle regeneration from 2010 to 2023 using bibliometric analyses, providing valuable insights into numerous future research directions.
Collapse
Affiliation(s)
- Nan Huang
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Intelligent Rehabilitation Technology Innovation Center, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Kang Zou
- Department of Critical Care Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Yanbiao Zhong
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Yun Luo
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Maoyuan Wang
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
| | - Li Xiao
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou City, Jiangxi Province, 341000, PR China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, Jiangxi Province, 341000, PR China
| |
Collapse
|
12
|
Hachemi Y, Perrin S, Ethel M, Julien A, Vettese J, Geisler B, Göritz C, Colnot C. Multimodal analyses of immune cells during bone repair identify macrophages as a therapeutic target in musculoskeletal trauma. Bone Res 2024; 12:56. [PMID: 39341816 PMCID: PMC11438896 DOI: 10.1038/s41413-024-00347-3] [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: 12/14/2023] [Revised: 04/04/2024] [Accepted: 05/23/2024] [Indexed: 10/01/2024] Open
Abstract
Musculoskeletal traumatic injuries (MTI) involve soft tissue lesions adjacent to a bone fracture leading to fibrous nonunion. The impact of MTI on the inflammatory response to fracture and on the immunomodulation of skeletal stem/progenitor cells (SSPCs) remains unknown. Here, we used single-nucleus transcriptomic analyses to describe the immune cell dynamics after bone fracture and identified distinct macrophage subsets with successive pro-inflammatory, pro-repair and anti-inflammatory profiles. Concurrently, SSPCs transition via a pro- and anti-inflammatory fibrogenic phase of differentiation prior to osteochondrogenic differentiation. In a preclinical MTI mouse model, the injury response of immune cells and SSPCs is disrupted leading to a prolonged pro-inflammatory phase and delayed resolution of inflammation. Macrophage depletion improves bone regeneration in MTI demonstrating macrophage involvement in fibrous nonunion. Finally, pharmacological inhibition of macrophages using the CSF1R inhibitor Pexidartinib ameliorates healing. These findings reveal the coordinated immune response of macrophages and skeletal stem/progenitor cells as a driver of bone healing and as a primary target for the treatment of trauma-associated fibrosis.
Collapse
Affiliation(s)
| | - Simon Perrin
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| | - Maria Ethel
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| | - Anais Julien
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Julia Vettese
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| | | | - Christian Göritz
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, Hong Kong
| | - Céline Colnot
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.
| |
Collapse
|
13
|
Xiong Y, Mi B, Liu G, Zhao Y. Microenvironment-sensitive nanozymes for tissue regeneration. Biomaterials 2024; 309:122585. [PMID: 38692147 DOI: 10.1016/j.biomaterials.2024.122585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
Tissue defect is one of the significant challenges encountered in clinical practice. Nanomaterials, including nanoparticles, nanofibers, and metal-organic frameworks, have demonstrated an extensive potential in tissue regeneration, offering a promising avenue for future clinical applications. Nonetheless, the intricate landscape of the inflammatory tissue microenvironment has engendered challenges to the efficacy of nanomaterial-based therapies. This quandary has spurred researchers to pivot towards advanced nanotechnological remedies for overcoming these therapeutic constraints. Among these solutions, microenvironment-sensitive nanozymes have emerged as a compelling instrument with the capacity to reshape the tissue microenvironment and enhance the intricate process of tissue regeneration. In this review, we summarize the microenvironmental characteristics of damaged tissues, offer insights into the rationale guiding the design and engineering of microenvironment-sensitive nanozymes, and explore the underlying mechanisms that underpin these nanozymes' responsiveness. This analysis includes their roles in orchestrating cellular signaling, modulating immune responses, and promoting the delicate process of tissue remodeling. Furthermore, we discuss the diverse applications of microenvironment-sensitive nanozymes in tissue regeneration, including bone, soft tissue, and cartilage regeneration. Finally, we shed our sights on envisioning the forthcoming milestones in this field, prospecting a future where microenvironment-sensitive nanozymes contribute significantly to the development of tissue regeneration and improved clinical outcomes.
Collapse
Affiliation(s)
- Yuan Xiong
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Bobin Mi
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore; Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore.
| |
Collapse
|
14
|
Li L, Li D, Zhu J, Wang Y, Zhao F, Cheng J, Tuan RS, Hu X, Ao Y. Downregulation of TGF-β1 in fibro-adipogenic progenitors initiates muscle ectopic mineralization. J Bone Miner Res 2024; 39:1147-1161. [PMID: 38896028 DOI: 10.1093/jbmr/zjae097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 05/29/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
In previous studies, we have demonstrated that stress response-induced high glucocorticoid levels could be the underlying cause of traumatic heterotopic ossification (HO), and we have developed a glucocorticoid-induced ectopic mineralization (EM) mouse model by systemic administration of a high dose of dexamethasone (DEX) to animals with muscle injury induced by cardiotoxin injection. In this model, dystrophic calcification (DC) developed into HO in a cell autonomous manner. However, it is not clear how DC is formed after DEX treatment. Therefore, in this study, we aimed to explore how glucocorticoids initiate muscle EM at a cellular and molecular level. We showed that DEX treatment inhibited inflammatory cell infiltration into injured muscle but inflammatory cytokine production in the muscle was significantly increased, suggesting that other non-inflammatory muscle cell types may regulate the inflammatory response and the muscle repair process. Accompanying this phenotype, transforming growth factor β1 (TGF-β1) expression in fibro-adipogenic progenitors (FAPs) was greatly downregulated. Since TGF-β1 is a strong immune suppressor and FAP's regulatory role has a large impact on muscle repair, we hypothesized that downregulation of TGF-β1 in FAPs after DEX treatment resulted in this hyperinflammatory state and subsequent failed muscle repair and EM formation. To test our hypothesis, we utilized a transgenic mouse model to specifically knockout Tgfb1 gene in PDGFRα-positive FAPs to investigate if the transgenic mice could recapitulate the phenotype that was induced by DEX treatment. Our results showed that the transgenic mice completely phenocopied this hyperinflammatory state and spontaneously developed EM following muscle injury. On the contrary, therapeutics that enhanced TGF-β1 signaling in FAPs inhibited the inflammatory response and attenuated muscle EM. In summary, these results indicate that FAPs-derived TGF-β1 is a key molecule in regulating muscle inflammatory response and subsequent EM, and that glucocorticoids exert their effect via downregulating TGF-β1 in FAPs.
Collapse
Affiliation(s)
- La Li
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Dai Li
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Jingxian Zhu
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Yiqun Wang
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Feng Zhao
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Jin Cheng
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Xiaoqing Hu
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Yingfang Ao
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| |
Collapse
|
15
|
Guan Y, Li X, Yang H, Xu S, Shi L, Liu Y, Kong L, Qin Y. Role and mechanism of IRF9 in promoting the progression of rheumatoid arthritis by regulating macrophage polarization via PSMA5. Heliyon 2024; 10:e35589. [PMID: 39170377 PMCID: PMC11336755 DOI: 10.1016/j.heliyon.2024.e35589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024] Open
Abstract
Aim To explore the mechanisms of IRF9 in the progression of rheumatoid arthritis(RA), and the effects of IRF9 on M1/M2 polarization. Methods RA dataset (GSE55457) was downloaded from GEO. Correlation analysis between IRF9 and its downstream target protein PSMA5 was performed using bioinformatics analysis. The M1/M2 cell ratio of peripheral blood mononuclear cells which from 20 healthy specimen and 40 RA patients was determined. The expression of IRF9 and PSMA5 was detected using qPCR and Western blot. Then, knockdown IRF9 in RAW264.7 cell line (sh-IRF9 RAW264.7) was constructed. The effect of sh-IRF9 RAW264.7 on RA was explored by constructing a CIA mouse model. Results IRF9 is upregulated in RA and is of good early screening effect. The results of pathway analysis showed that IRF9 targets and regulates the PSMA5 signaling pathway. IRF9 and PSMA5 were significantly elevated in RA patients, M1/M2 ratio was also increased. The effects of IRF9 on RAW264.7 macrophages were deeply explored in vitro, revealing that knockdown of IRF9 suppressed PSMA5, M1/M2 ratio and the secretion of pro-inflammatory factor in RAW264.7. In mouse in vivo experiments, sh-IRF9 RAW264.7 cells were found to modulate RA by downregulating PSMA5, modulating the M1/M2 ratio through enhancing the anti-inflammatory factor, and suppressing the pro-inflammatory factor. Conclusion IRF9 promoted the progression of RA via regulating macrophage polarization through PSMA5.
Collapse
Affiliation(s)
- Yue Guan
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Xin Li
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Hemin Yang
- Central Laboratory, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Siyu Xu
- Inspection Center, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Lidong Shi
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Yangyang Liu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Lingdan Kong
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Ying Qin
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
16
|
Dai Q, Wan C, Xu Y, Fei K, Olivere LA, Garrett B, Akers L, Peters D, Otto J, Kontos CD, Ji Z, Diao Y, Southerland KW. Vcam1+ Fibro-adipogenic Progenitors Mark Fatty Infiltration in Chronic Limb Threatening Ischemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602430. [PMID: 39026697 PMCID: PMC11257459 DOI: 10.1101/2024.07.08.602430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Skeletal muscle health and function is a critical determinant of clinical outcomes in patients with peripheral arterial disease (PAD). Herein, we identify fatty infiltration, the ectopic deposition of adipocytes in skeletal muscle, as a histological hallmark of end-stage PAD, also known as chronic limb threatening ischemia (CLTI). Leveraging single cell transcriptome mapping in mouse models of PAD, we identify a pro-adipogenic mesenchymal stromal cell population marked by expression of Vcam1 (termed Vcam1+ FAPs) that expands in the ischemic limb. Mechanistically, we identify Sfrp1 and Nr3c1 as regulators of Vcam1+ FAP adipogenic differentiation. Loss of Sfrp1 and Nr3c1 impair Vcam1+ FAP differentiation into adipocytes in vitro. Finally, we show that Vcam1+ FAPs are enriched in human CLTI patients. Collectively, our results identify a pro-adipogenic FAP subpopulation in CLTI patients and provide a potential therapeutic target for muscle regeneration in PAD.
Collapse
Affiliation(s)
- Qunsheng Dai
- Division of Vascular and Endovascular Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Changxin Wan
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Yueyuan Xu
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - Kaileen Fei
- Duke University School of Medicine, Duke University, Durham, NC, USA
| | - Lindsey A Olivere
- Division of Vascular Surgery, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Brianna Garrett
- Division of Vascular and Endovascular Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Leo Akers
- Division of Vascular and Endovascular Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Derek Peters
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - James Otto
- Division of Vascular and Endovascular Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Christopher D Kontos
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Zhiceng Ji
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Yarui Diao
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC, USA
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Kevin W Southerland
- Division of Vascular and Endovascular Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
17
|
Rodríguez C, Timóteo-Ferreira F, Minchiotti G, Brunelli S, Guardiola O. Cellular interactions and microenvironment dynamics in skeletal muscle regeneration and disease. Front Cell Dev Biol 2024; 12:1385399. [PMID: 38840849 PMCID: PMC11150574 DOI: 10.3389/fcell.2024.1385399] [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: 02/12/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Skeletal muscle regeneration relies on the intricate interplay of various cell populations within the muscle niche-an environment crucial for regulating the behavior of muscle stem cells (MuSCs) and ensuring postnatal tissue maintenance and regeneration. This review delves into the dynamic interactions among key players of this process, including MuSCs, macrophages (MPs), fibro-adipogenic progenitors (FAPs), endothelial cells (ECs), and pericytes (PCs), each assuming pivotal roles in orchestrating homeostasis and regeneration. Dysfunctions in these interactions can lead not only to pathological conditions but also exacerbate muscular dystrophies. The exploration of cellular and molecular crosstalk among these populations in both physiological and dystrophic conditions provides insights into the multifaceted communication networks governing muscle regeneration. Furthermore, this review discusses emerging strategies to modulate the muscle-regenerating niche, presenting a comprehensive overview of current understanding and innovative approaches.
Collapse
Affiliation(s)
- Cristina Rodríguez
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso”, CNR, Naples, Italy
| | | | - Gabriella Minchiotti
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso”, CNR, Naples, Italy
| | - Silvia Brunelli
- School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Ombretta Guardiola
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati-Traverso”, CNR, Naples, Italy
| |
Collapse
|
18
|
Hachemi Y, Perrin S, Ethel M, Julien A, Vettese J, Geisler B, Göritz C, Colnot C. Multimodal analyses of immune cells during bone repair identify macrophages as a therapeutic target in musculoskeletal trauma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.591608. [PMID: 38746344 PMCID: PMC11092472 DOI: 10.1101/2024.04.29.591608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Musculoskeletal traumatic injuries (MTI) involve soft tissue lesions adjacent to a bone fracture leading to fibrous nonunion. The impact of MTI on the inflammatory response to fracture and on the immunomodulation of skeletal stem/progenitor cells (SSPCs) remains unknown. Here, we used single cell transcriptomic analyses to describe the immune cell dynamics after bone fracture and identified distinct macrophage subsets with successive pro-inflammatory, pro-repair and anti-inflammatory profiles. Concurrently, SSPCs transition via a pro- and anti-inflammatory fibrogenic phase of differentiation prior to osteochondrogenic differentiation. In a preclinical MTI mouse model, the injury response of immune cells and SSPCs is disrupted leading to a prolonged pro-inflammatory phase and delayed resolution of inflammation. Macrophage depletion improves bone regeneration in MTI demonstrating macrophage involvement in fibrous nonunion. Finally, pharmacological inhibition of macrophages using the CSF1R inhibitor Pexidartinib ameliorates healing. These findings reveal the coordinated immune response of macrophages and skeletal stem/progenitor cells as driver of bone healing and as a primary target for the treatment of trauma-associated fibrosis. Summary Hachemi et al. report the immune cell atlas of bone repair revealing macrophages as pro-fibrotic regulators and a therapeutic target for musculoskeletal regeneration. Genetic depletion or pharmacological inhibition of macrophages improves bone healing in musculoskeletal trauma.
Collapse
|
19
|
King PH. Skeletal muscle as a molecular and cellular biomarker of disease progression in amyotrophic lateral sclerosis: a narrative review. Neural Regen Res 2024; 19:747-753. [PMID: 37843208 PMCID: PMC10664124 DOI: 10.4103/1673-5374.382226] [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: 05/07/2023] [Revised: 06/27/2023] [Accepted: 07/19/2023] [Indexed: 10/17/2023] Open
Abstract
Amyotrophic lateral sclerosis is a fatal multisystemic neurodegenerative disease with motor neurons being a primary target. Although progressive weakness is a hallmark feature of amyotrophic lateral sclerosis, there is considerable heterogeneity, including clinical presentation, progression, and the underlying triggers for disease initiation. Based on longitudinal studies with families harboring amyotrophic lateral sclerosis-associated gene mutations, it has become apparent that overt disease is preceded by a prodromal phase, possibly in years, where compensatory mechanisms delay symptom onset. Since 85-90% of amyotrophic lateral sclerosis is sporadic, there is a strong need for identifying biomarkers that can detect this prodromal phase as motor neurons have limited capacity for regeneration. Current Food and Drug Administration-approved therapies work by slowing the degenerative process and are most effective early in the disease. Skeletal muscle, including the neuromuscular junction, manifests abnormalities at the earliest stages of the disease, before motor neuron loss, making it a promising source for identifying biomarkers of the prodromal phase. The accessibility of muscle through biopsy provides a lens into the distal motor system at earlier stages and in real time. The advent of "omics" technology has led to the identification of numerous dysregulated molecules in amyotrophic lateral sclerosis muscle, ranging from coding and non-coding RNAs to proteins and metabolites. This technology has opened the door for identifying biomarkers of disease activity and providing insight into disease mechanisms. A major challenge is correlating the myriad of dysregulated molecules with clinical or histological progression and understanding their relevance to presymptomatic phases of disease. There are two major goals of this review. The first is to summarize some of the biomarkers identified in human amyotrophic lateral sclerosis muscle that have a clinicopathological correlation with disease activity, evidence of a similar dysregulation in the SOD1G93A mouse during presymptomatic stages, and evidence of progressive change during disease progression. The second goal is to review the molecular pathways these biomarkers reflect and their potential role in mitigating or promoting disease progression, and as such, their potential as therapeutic targets in amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Peter H. King
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, AL, USA; Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA
| |
Collapse
|
20
|
Liu K, Wang Y, Shao W, Tang C, Zhao S, Xu J, Xu P, Cheng Q, Huang S, Ji P, Qiu S. Unveiling the oncogenic role of CLDN11-secreting fibroblasts in gastric cancer peritoneal metastasis through single-cell sequencing and experimental approaches. Int Immunopharmacol 2024; 129:111647. [PMID: 38335659 DOI: 10.1016/j.intimp.2024.111647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/14/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Fibroblasts are necessary to the progression of cancer. However, the role of fibroblasts in peritoneal metastasis (PM) of gastric cancer (GC) remains elusive. In this study, we would explore the role of fibroblasts mediated cell interaction in PM of GC. METHODS Single-cell sequencing data from public database GSE183904 was used to explore the specific fibroblast cluster. Fibroblasts were extracted from PM and GC tissues. The expression level of CXCR7 was verified by western blot, immunohistochemistry. The role of CLDN11 was investigate through in vitro and in vivo study. Multiple immunohistochemistry was used to characterize the tumor microenvironment. RESULTS CXCR7-positive fibroblasts were significantly enriched in PM of GC. CXCR7 could promote the expression of CLDN11 through activation of the AKT pathway in fibroblasts. Fibroblasts promote the GC proliferation and peritoneal metastasis by secreting CLDN11 in vitro and in vivo. Furthermore, it was revealed that CXCR7-positive fibroblasts were significantly associated with M2-type macrophages infiltration in tissues. CONCLUSION CXCR7-positive fibroblasts play an essential role in PM of GC via CLDN11. Therapy targeting CXCR7-positive fibroblasts or CLDN11 may be helpful in the treatment of GC with PM.
Collapse
Affiliation(s)
- Kanghui Liu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yanjuan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Wenwen Shao
- Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Chong Tang
- Department of General Surgery, Nantong First People's Hospital, Nantong, Jiangsu Province, China
| | - Siguo Zhao
- Department of Clinical Medicine, The First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Jiafeng Xu
- Department of Clinical Medicine, The First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Peng Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Quan Cheng
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shansong Huang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Peicheng Ji
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shengkui Qiu
- Department of General Surgery, Nantong First People's Hospital, Nantong, Jiangsu Province, China.
| |
Collapse
|
21
|
Shao C, Xu H, Sun X, Pan Y, Liang X, Huang J, He Y, Guo W, Ye L, Zhang J. Jiawei Taohe Chengqi decoction inhibition of the notch signal pathway affects macrophage reprogramming to inhibit HSCs activation for the treatment of hepatic fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117486. [PMID: 38030027 DOI: 10.1016/j.jep.2023.117486] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/12/2023] [Accepted: 11/20/2023] [Indexed: 12/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiawei Taohe Chengqi Tang (JTCD) is a modified formulation of Traditional Chinese Medicine (TCM) known as Taohe Chengqi Decoction, which has been described in the ancient TCM literature "Treatise on Febrile Diseases". As a formula that can activate blood circulation and eliminate blood stasis and regulate Yin and Yang in traditional Chinese medicine applications, JTCD has been reported to be effective in the treatment of chronic liver disease and hepatic fibrosis (HF). AIM OF STUDY The current study aimed to evaluate the effectiveness of JTCD in modulating hepatic macrophages by regulating the Notch signal pathway, and to further investigate the mechanisms underlying macrophage reprogramming that leads to HF. MATERIALS AND METHODS Molecular assays were performed using in vitro cultures of human mononuclear THP-1 cells and human-derived hepatic stellate cells LX-2. CCl4-induced mice were utilized as an in vivo model to simulate HF. RESULTS Our results demonstrated that JTCD exhibited dual effects by inhibiting hepatic stellate cell (HSCs) activation and modulating the polarisation of macrophages towards the M2 phenotype while decreasing the M1 phenotype. Network pharmacological analyses and molecular docking studies revealed that the Notch signal pathway was significantly enriched and played a crucial role in the therapeutic response of JTCD against HF. Moreover, through the establishment of a co-culture model, we validated that JTCD inhibited the Notch signal pathway in macrophages, leading to alterations in macrophage reprogramming, subsequent inhibition of HSC activation, and ultimately exerting anti-HF effects. CONCLUSION In conclusion, our findings provide solid evidence for JTCD in treating HF, as it suppresses the Notch signal pathway in macrophages, regulates macrophage reprogramming, and inhibits HSC activation.
Collapse
Affiliation(s)
- Chang Shao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Huihui Xu
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Xiguang Sun
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Yun Pan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Xiaofan Liang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Jiaxin Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Yi He
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Wenqin Guo
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Linmao Ye
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Junjie Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
22
|
Ngo TB, Josyula A, DeStefano S, Fertil D, Faust M, Lokwani R, Sadtler K. Intersection of Immunity, Metabolism, and Muscle Regeneration in an Autoimmune-Prone MRL Mouse Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306961. [PMID: 38192168 PMCID: PMC10953568 DOI: 10.1002/advs.202306961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/07/2023] [Indexed: 01/10/2024]
Abstract
Due to the limited capacity of mammals to regenerate complex tissues, researchers have worked to understand the mechanisms of tissue regeneration in organisms that maintain that capacity. One example is the MRL/MpJ mouse strain with unique regenerative capacity in ear pinnae that is absent from other strains, such as the common C57BL/6 strain. The MRL/MpJ mouse has also been associated with an autoimmune phenotype even in the absence of the mutant Fas gene described in its parent strain MRL/lpr. Due to these findings, the differences between the responses of MRL/MpJ versus C57BL/6 strain are evaluated in volumetric muscle injury and subsequent material implantation. One salient feature of the MRL/MpJ response to injury is robust adipogenesis within the muscle. This is associated with a decrease in M2-like polarization in response to biologically derived extracellular matrix scaffolds. In pro-fibrotic materials, such as polyethylene, there are fewer foreign body giant cells in the MRL/MpJ mice. As there are reports of both positive and negative influences of adipose tissue and adipogenesis on wound healing, this model can provide an important lens to investigate the interplay between stem cells, adipose tissue, and immune responses in trauma and material implantation.
Collapse
Affiliation(s)
- Tran B. Ngo
- Section on ImmunoengineeringCenter for Biomedical Engineering and Technology AccelerationNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20814USA
| | - Aditya Josyula
- Section on ImmunoengineeringCenter for Biomedical Engineering and Technology AccelerationNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20814USA
| | - Sabrina DeStefano
- Section on ImmunoengineeringCenter for Biomedical Engineering and Technology AccelerationNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20814USA
| | - Daphna Fertil
- Section on ImmunoengineeringCenter for Biomedical Engineering and Technology AccelerationNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20814USA
| | - Mondreakest Faust
- Section on ImmunoengineeringCenter for Biomedical Engineering and Technology AccelerationNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20814USA
| | - Ravi Lokwani
- Section on ImmunoengineeringCenter for Biomedical Engineering and Technology AccelerationNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20814USA
| | - Kaitlyn Sadtler
- Section on ImmunoengineeringCenter for Biomedical Engineering and Technology AccelerationNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20814USA
| |
Collapse
|
23
|
Wei D, Fan J, Yan J, Liu C, Cao J, Xu C, Sun Y, Xiao H. Nuclear-Targeting Lipid Pt IV Prodrug Amphiphile Cooperates with siRNA for Enhanced Cancer Immunochemotherapy by Amplifying Pt-DNA Adducts and Reducing Phosphatidylserine Exposure. J Am Chem Soc 2024; 146:1185-1195. [PMID: 38148611 DOI: 10.1021/jacs.3c12706] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Patients treated with Pt-based anticancer drugs (PtII) often experience severe side effects and are susceptible to cancer recurrence due to the limited bioavailability of PtII and tumor-induced immunosuppression. The exposure of phosphatidylserine on the cell's outer surface induced by PtII results in profound immunosuppression through the binding of phosphatidylserine to its receptors on immune cells. Here, we report a novel approach for enhanced cancer chemoimmunotherapy, where a novel nuclear-targeting lipid PtIV prodrug amphiphile was used to deliver a small interfering RNA (siXkr8) to simultaneously amplify Pt-DNA adducts and reduce the level of exposure of phosphatidylserine. This drug delivery vehicle is engineered by integrating the PtIV prodrug with self-assembly performance and siXkr8 into a lipid nanoparticle, which shows tumor accumulation, cancer cell nucleus targeting, and activatable in a reduced microenvironment. It is demonstrated that nuclear-targeting lipid PtIV prodrug increases the DNA cross-linking, resulting in increased Pt-DNA adduct formation. The synergistic effects of the PtIV prodrug and siXkr8 contribute to the improvement of the tumor immune microenvironment. Consequently, the increased Pt-DNA adducts and immunogenicity effectively inhibit primary tumor growth and prevent tumor recurrence. These results underscore the potential of utilizing the nuclear-targeting lipid PtIV prodrug amphiphile to enhance Pt-DNA adduct formation and employing siXkr8 to alleviate immunosuppression during chemotherapy.
Collapse
Affiliation(s)
- Dengshuai Wei
- Department of Pharmaceutics, School of Pharmacy and Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Junning Fan
- Department of Pharmaceutics, School of Pharmacy and Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Jianqin Yan
- Department of Pharmaceutics, School of Pharmacy and Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Chaolong Liu
- Department of Pharmaceutics, School of Pharmacy and Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Jie Cao
- Department of Pharmaceutics, School of Pharmacy and Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Chun Xu
- School of Dentistry, The University of Queensland, Brisbane 4006, Australia
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy and Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
24
|
Gao Y, Qi Y, Shen Y, Zhang Y, Wang D, Su M, Liu X, Wang A, Zhang W, He C, Yang J, Dai M, Wang H, Cai H. Signatures of tumor-associated macrophages correlate with treatment response in ovarian cancer patients. Aging (Albany NY) 2024; 16:207-225. [PMID: 38175687 PMCID: PMC10817412 DOI: 10.18632/aging.205362] [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: 06/27/2023] [Accepted: 11/02/2023] [Indexed: 01/05/2024]
Abstract
Ovarian cancer (OC) ranks as the second leading cause of death among gynecological cancers. Numerous studies have indicated a correlation between the tumor microenvironment (TME) and the clinical response to treatment in OC patients. Tumor-associated macrophages (TAMs), a crucial component of the TME, exert influence on invasion, metastasis, and recurrence in OC patients. To delve deeper into the role of TAMs in OC, this study conducted an extensive analysis of single-cell data from OC patients. The aim is to develop a new risk score (RS) to characterize the response to treatment in OC patients to inform clinical treatment. We first identified TAM-associated genes (TAMGs) in OC patients and examined the protein and mRNA expression levels of TAMGs by Western blot and PCR experiments. Additionally, a scoring system for TAMGs was constructed, successfully categorizing patients into high and low RS subgroups. Remarkably, significant disparities were observed in immune cell infiltration and immunotherapy response between the high and low RS subgroups. The findings revealed that patients in the high RS group had a poorer prognosis but displayed greater sensitivity to immunotherapy. Another important finding was that patients in the high RS subgroup had a higher IC50 for chemotherapeutic agents. Furthermore, further experimental investigations led to the discovery that THEMIS2 could serve as a potential target in OC patients and is associated with EMT (epithelial-mesenchymal transition). Overall, the TAMGs-based scoring system holds promise for screening patients who would benefit from therapy and provides valuable information for the clinical treatment of OC.
Collapse
Affiliation(s)
- Yang Gao
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Yuwen Qi
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Yin Shen
- Department of Integrative Ultrasound Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yaxing Zhang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Dandan Wang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Min Su
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Xuelian Liu
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Anjin Wang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Wenwen Zhang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Can He
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Junyuan Yang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Mengyuan Dai
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Hua Wang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Hongbing Cai
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| |
Collapse
|
25
|
Jia D, He Y, Zhang Y. Long Non-coding RNAs Regulating Macrophage Polarization in Liver Cancer. Curr Pharm Des 2024; 30:2120-2128. [PMID: 38859791 DOI: 10.2174/0113816128311861240523075218] [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: 03/17/2024] [Accepted: 04/24/2024] [Indexed: 06/12/2024]
Abstract
Primary liver cancer is the second leading cause of cancer-related death worldwide. At present, liver cancer is often in an advanced stage once diagnosed, and treatment effects are generally poor. Therefore, there is an urgent need for other powerful treatments. Macrophages are an important component of the tumor microenvironment, and macrophage polarization is crucial to tumor proliferation and differentiation. Regulatory interactions between macrophage subtypes, such as M1 and M2, lead to a number of clinical outcomes, including tumor progression and metastasis. So, it is important to study the drivers of this process. Long non-coding RNA has been widely proven to be of great value in the early diagnosis and treatment of tumors. Many studies have shown that long non-coding RNA participates in macrophage polarization through its ability to drive M1 or M2 polarization, thereby participating in the occurrence and development of liver cancer. In this article, we systematically elaborated on the long non-coding RNAs involved in the polarization of liver cancer macrophages, hoping to provide a new idea for the early diagnosis and treatment of liver cancer. Liver cancer- related studies were retrieved from PubMed. Based on our identification of lncRNA and macrophage polarization as powerful therapies for liver cancer, we analyzed research articles in the PubMed system in the last ten years on the crosstalk between lncRNA and macrophage polarization. By targeting M1/M2 macrophage polarization, lncRNA may promote or suppress liver cancer, and the references are determined primarily by the article's impact factor. Consequently, the specific mechanism of action between lncRNA and M1/M2 macrophage polarization was explored, along with the role of their crosstalk in the occurrence, proliferation, and metastasis of liver cancer. LncRNA is bidirectionally expressed in liver cancer and can target macrophage polarization to regulate tumor behavior. LncRNA mainly functions as ceRNA and can participate in the crosstalk between liver cancer cells and macrophages through extracellular vesicles. LncRNA can potentially participate in the immunotherapy of liver cancer by targeting macrophages and becoming a new biomolecular marker of liver cancer.
Collapse
Affiliation(s)
- Dengke Jia
- Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Yaping He
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Yawu Zhang
- Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Hepato-Biliary-Pancreatic Institute, Lanzhou University Second Hospital, Lanzhou 730000, China
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China
| |
Collapse
|
26
|
Luo W, Zhou Y, Tang Q, Wang Y, Liu Y, Ai L. Downhill running and caloric restriction attenuate insulin resistance associated skeletal muscle atrophy via the promotion of M2-like macrophages through TRIB3-AKT pathway. Free Radic Biol Med 2024; 210:271-285. [PMID: 38036069 DOI: 10.1016/j.freeradbiomed.2023.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/11/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUD Downhill running has recently become a promising exercise modality for metabolic syndrome, but the effect and precise mechanism of downhill running training on insulin resistance (IR) induced skeletal muscle atrophy remains unclear. The current study aimed to explore the benefits of downhill running training accompanied by a low-fat diet on skeletal muscle atrophy in IR mice and its possible mechanisms. METHODS For in vivo study, high fat diet (HFD) -induced IR mice were submitted to the downhill running training or/and caloric restriction for 8 weeks. In vitro study was performed using co-cultured RAW264.7 macrophages and C2C12 myoblasts model. Glucose tolerance test (GTT), insulin tolerance test (ITT), immunofluorescence staining, Western blot analysis, hematoxylin and eosin (H&E) staining, enzyme-linked immunosorbent assay (ELISA), Cell counting kit-8 (CCK-8) assays and glucose uptake assays were employed to explore the benefits and possible mechanisms of downhill running training accompanied by a low-fat diet on IR mice. RESULTS Our data revealed that HFD induces IR, which leading to skeletal muscle atrophy. Downhill running accompanied by caloric restriction mitigated HFD-induced IR and improve skeletal muscle atrophy. Further study suggested that descended TRIB3 mediated the favorable impact of downhill running on IR induced skeletal muscle atrophy by suppressing M1-like macrophages and promoting M2-like macrophages. Macrophages-specific knockdown of TRIB3 exerted similar effects on the macrophage polarization and IR related myogenesis to downhill running training accompanied by caloric restriction. In contrast, macrophages-specific overexpression of TRIB3 descended phosphorylation of AKT, further activated M1-like macrophages and aggravated IR related inhibition of myogenesis. CONCLUSIONS This finding demonstrated the beneficial effects of downhill running training and caloric restriction on IR related skeletal muscle atrophy by promoting M2-like macrophages through TRIB3-AKT pathway.
Collapse
Affiliation(s)
- Wei Luo
- Department of Sports and Health Sciences, Nanjing Sport Institute, Nanjing, China
| | - Yue Zhou
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Qiang Tang
- Department of Sports and Health Sciences, Nanjing Sport Institute, Nanjing, China
| | - Yuhang Wang
- Department of Sports and Health Sciences, Nanjing Sport Institute, Nanjing, China
| | - Yansong Liu
- Department of Sports and Health Sciences, Nanjing Sport Institute, Nanjing, China
| | - Lei Ai
- Jiangsu Research Institute of Sports Science, Nanjing, China.
| |
Collapse
|
27
|
Lequain H, Dégletagne C, Streichenberger N, Valantin J, Simonet T, Schaeffer L, Sève P, Leblanc P. Spatial Transcriptomics Reveals Signatures of Histopathological Changes in Muscular Sarcoidosis. Cells 2023; 12:2747. [PMID: 38067175 PMCID: PMC10706822 DOI: 10.3390/cells12232747] [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: 11/09/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Sarcoidosis is a multisystemic disease characterized by non-caseating granuloma infiltrating various organs. The form with symptomatic muscular involvement is called muscular sarcoidosis. The impact of immune cells composing the granuloma on the skeletal muscle is misunderstood. Here, we investigated the granuloma-skeletal muscle interactions through spatial transcriptomics on two patients affected by muscular sarcoidosis. Five major transcriptomic clusters corresponding to perigranuloma, granuloma, and three successive muscle tissue areas (proximal, intermediate, and distal) around the granuloma were identified. Analyses revealed upregulated pathways in the granuloma corresponding to the activation of T-lymphocytes and monocytes/macrophages cytokines, the upregulation of extracellular matrix signatures, and the induction of the TGF-β signaling in the perigranuloma. A comparison between the proximal and distal muscles to the granuloma revealed an inverse correlation between the distance to the granuloma and the upregulation of cellular response to interferon-γ/α, TNF-α, IL-1,4,6, fibroblast proliferation, epithelial to mesenchymal cell transition, and the downregulation of muscle gene expression. These data shed light on the intercommunications between granulomas and the muscle tissue and provide pathophysiological mechanisms by showing that granuloma immune cells have a direct impact on proximal muscle tissue by promoting its progressive replacement by fibrosis via the expression of pro-inflammatory and profibrosing signatures. These data could possibly explain the evolution towards a state of disability for some patients.
Collapse
Affiliation(s)
- Hippolyte Lequain
- Département de Médecine Interne, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, 69004 Lyon, France;
- Institut NeuroMyoGène INMG-PGNM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, Inserm U1315, Faculté de Médecine Rockefeller, Université Claude Bernard UCBL-Lyon 1, 69008 Lyon, France; (N.S.); (T.S.)
| | - Cyril Dégletagne
- CRCL Core Facilities, Centre de Recherche en Cancérologie de Lyon (CRCL) INSERM U1052-CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon1, Centre Léon Bérard, 69008 Lyon, France; (C.D.); (J.V.)
| | - Nathalie Streichenberger
- Institut NeuroMyoGène INMG-PGNM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, Inserm U1315, Faculté de Médecine Rockefeller, Université Claude Bernard UCBL-Lyon 1, 69008 Lyon, France; (N.S.); (T.S.)
- Service d’Anatomopathologie, Centre de Biologie et Pathologie Est (CBPE), Hospices Civils de Lyon, 69500 Bron, France
| | - Julie Valantin
- CRCL Core Facilities, Centre de Recherche en Cancérologie de Lyon (CRCL) INSERM U1052-CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon1, Centre Léon Bérard, 69008 Lyon, France; (C.D.); (J.V.)
| | - Thomas Simonet
- Institut NeuroMyoGène INMG-PGNM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, Inserm U1315, Faculté de Médecine Rockefeller, Université Claude Bernard UCBL-Lyon 1, 69008 Lyon, France; (N.S.); (T.S.)
| | - Laurent Schaeffer
- Institut NeuroMyoGène INMG-PGNM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, Inserm U1315, Faculté de Médecine Rockefeller, Université Claude Bernard UCBL-Lyon 1, 69008 Lyon, France; (N.S.); (T.S.)
- Centre de Biotechnologie Cellulaire, CHU de Lyon—HCL Groupement Est, 69677 Bron, France
| | - Pascal Sève
- Département de Médecine Interne, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, 69004 Lyon, France;
- Pôle IMER, HESPER EA 7425, 69002 Lyon, France
| | - Pascal Leblanc
- Institut NeuroMyoGène INMG-PGNM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, Inserm U1315, Faculté de Médecine Rockefeller, Université Claude Bernard UCBL-Lyon 1, 69008 Lyon, France; (N.S.); (T.S.)
| |
Collapse
|
28
|
Ngo TB, Josyula A, DeStefano S, Fertil D, Faust M, Lokwani R, Sadtler K. Ectopic adipogenesis in response to injury and material implantation in an autoimmune mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.05.561105. [PMID: 37986843 PMCID: PMC10659416 DOI: 10.1101/2023.10.05.561105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Due to the limited capacity of mammals to regenerate complex tissues, researchers have worked to understand the mechanisms of tissue regeneration in organisms that maintain that capacity. One example is the MRL/MpJ mouse strain with unique regenerative capacity in ear pinnae that is absent from other strains, such as the common C57BL/6 strain. The MRL/MpJ mouse has also been associated with an autoimmune phenotype even in the absence of the mutant Fas gene described in its parent strain MRL/lpr. Due to these findings, we evaluated the differences between the responses of MRL/MpJ versus C57BL/6 strain in traumatic muscle injury and subsequent material implantation. One salient feature of the MRL/MpJ response to injury was a robust adipogenesis within the muscle. This was associated with a decrease in M2-like polarization in response to biologically derived extracellular matrix scaffolds. In pro-fibrotic materials, such as polyethylene, there were fewer foreign body giant cells in the MRL/MpJ mice. As there are reports of both positive and negative influences of adipose tissue and adipogenesis on wound healing, this model could provide an important lens to investigate the interplay between stem cells, adipose tissue, and immune responses in trauma and materials implantation.
Collapse
Affiliation(s)
- Tran B. Ngo
- Section on Immunoengineering, Center for Biomedical Engineering and Technology Acceleration, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda MD 20814
| | - Aditya Josyula
- Section on Immunoengineering, Center for Biomedical Engineering and Technology Acceleration, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda MD 20814
| | - Sabrina DeStefano
- Section on Immunoengineering, Center for Biomedical Engineering and Technology Acceleration, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda MD 20814
| | - Daphna Fertil
- Section on Immunoengineering, Center for Biomedical Engineering and Technology Acceleration, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda MD 20814
| | - Mondreakest Faust
- Section on Immunoengineering, Center for Biomedical Engineering and Technology Acceleration, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda MD 20814
| | - Ravi Lokwani
- Section on Immunoengineering, Center for Biomedical Engineering and Technology Acceleration, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda MD 20814
| | - Kaitlyn Sadtler
- Section on Immunoengineering, Center for Biomedical Engineering and Technology Acceleration, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda MD 20814
| |
Collapse
|
29
|
Gregg SR, Barshick MR, Johnson SE. Intravenous Injection of Sodium Hyaluronate Diminishes Basal Inflammatory Gene Expression in Equine Skeletal Muscle. Animals (Basel) 2023; 13:3030. [PMID: 37835636 PMCID: PMC10571686 DOI: 10.3390/ani13193030] [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: 08/18/2023] [Revised: 09/08/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Following strenuous exercise, skeletal muscle experiences an acute inflammatory state that initiates the repair process. Systemic hyaluronic acid (HA) is injected to horses routinely as a joint anti-inflammatory. To gain insight into the effects of HA on skeletal muscle, adult Thoroughbred geldings (n = 6) were injected with a commercial HA product weekly for 3 weeks prior to performing a submaximal exercise test. Gluteal muscle (GM) biopsies were obtained before and 1 h after exercise for gene expression analysis and HA localization. The results from RNA sequencing demonstrate differences in gene expression between non-injected controls (CON; n = 6) and HA horses. Prior to exercise, HA horses contained fewer (p < 0.05) transcripts associated with leukocyte activity and cytokine production than CON. The performance of exercise resulted in the upregulation (p < 0.05) of several cytokine genes and their signaling intermediates, indicating that HA does not suppress the normal inflammatory response to exercise. The transcript abundance for marker genes of neutrophils (NCF2) and macrophages (CD163) was greater (p < 0.05) post-exercise and was unaffected by HA injection. The anti-inflammatory effects of HA on muscle are indirect as no differences (p > 0.05) in the relative amount of the macromolecule was observed between the CON and HA fiber extracellular matrix (ECM). However, exercise tended (p = 0.10) to cause an increase in ECM size suggestive of muscle damage and remodeling. The finding was supported by the increased (p < 0.05) expression of CTGF, TGFβ1, MMP9, TIMP4 and Col4A1. Collectively, the results validate HA as an anti-inflammatory aid that does not disrupt the normal post-exercise muscle repair process.
Collapse
Affiliation(s)
| | | | - Sally E. Johnson
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA; (S.R.G.); (M.R.B.)
| |
Collapse
|
30
|
Coulis G, Jaime D, Guerrero-Juarez C, Kastenschmidt JM, Farahat PK, Nguyen Q, Pervolarakis N, McLinden K, Thurlow L, Movahedi S, Hughes BS, Duarte J, Sorn A, Montoya E, Mozaffar I, Dragan M, Othy S, Joshi T, Hans CP, Kimonis V, MacLean AL, Nie Q, Wallace LM, Harper SQ, Mozaffar T, Hogarth MW, Bhattacharya S, Jaiswal JK, Golann DR, Su Q, Kessenbrock K, Stec M, Spencer MJ, Zamudio JR, Villalta SA. Single-cell and spatial transcriptomics identify a macrophage population associated with skeletal muscle fibrosis. SCIENCE ADVANCES 2023; 9:eadd9984. [PMID: 37418531 PMCID: PMC10328414 DOI: 10.1126/sciadv.add9984] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 06/05/2023] [Indexed: 07/09/2023]
Abstract
Macrophages are essential for skeletal muscle homeostasis, but how their dysregulation contributes to the development of fibrosis in muscle disease remains unclear. Here, we used single-cell transcriptomics to determine the molecular attributes of dystrophic and healthy muscle macrophages. We identified six clusters and unexpectedly found that none corresponded to traditional definitions of M1 or M2 macrophages. Rather, the predominant macrophage signature in dystrophic muscle was characterized by high expression of fibrotic factors, galectin-3 (gal-3) and osteopontin (Spp1). Spatial transcriptomics, computational inferences of intercellular communication, and in vitro assays indicated that macrophage-derived Spp1 regulates stromal progenitor differentiation. Gal-3+ macrophages were chronically activated in dystrophic muscle, and adoptive transfer assays showed that the gal-3+ phenotype was the dominant molecular program induced within the dystrophic milieu. Gal-3+ macrophages were also elevated in multiple human myopathies. These studies advance our understanding of macrophages in muscular dystrophy by defining their transcriptional programs and reveal Spp1 as a major regulator of macrophage and stromal progenitor interactions.
Collapse
Affiliation(s)
- Gerald Coulis
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Diego Jaime
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | | | - Jenna M. Kastenschmidt
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Philip K. Farahat
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Quy Nguyen
- Department of Biological Chemistry, University of California Irvine, Irvine, CA USA
| | | | - Katherine McLinden
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Lauren Thurlow
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Saba Movahedi
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Brandon S. Hughes
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Jorge Duarte
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Andrew Sorn
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Elizabeth Montoya
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Izza Mozaffar
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
| | - Morgan Dragan
- Department of Biological Chemistry, University of California Irvine, Irvine, CA USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, MO, USA
| | - Chetan P. Hans
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO USA
| | - Virginia Kimonis
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Adam L. MacLean
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Qing Nie
- Department of Mathematics, Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Lindsay M. Wallace
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Scott Q. Harper
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Tahseen Mozaffar
- Department of Neurology, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| | - Marshall W. Hogarth
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Surajit Bhattacharya
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | - Jyoti K. Jaiswal
- Children’s National Hospital, Research Center for Genetic Medicine, Washington, DC, USA
| | | | - Qi Su
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California Irvine, Irvine, CA USA
| | - Michael Stec
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Melissa J. Spencer
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Jesse R. Zamudio
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - S. Armando Villalta
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
31
|
Cui CY, Ferrucci L, Gorospe M. Macrophage Involvement in Aging-Associated Skeletal Muscle Regeneration. Cells 2023; 12:1214. [PMID: 37174614 PMCID: PMC10177543 DOI: 10.3390/cells12091214] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
The skeletal muscle is a dynamic organ composed of contractile muscle fibers, connective tissues, blood vessels and nerve endings. Its main function is to provide motility to the body, but it is also deeply involved in systemic metabolism and thermoregulation. The skeletal muscle frequently encounters microinjury or trauma, which is primarily repaired by the coordinated actions of muscle stem cells (satellite cells, SCs), fibro-adipogenic progenitors (FAPs), and multiple immune cells, particularly macrophages. During aging, however, the capacity of skeletal muscle to repair and regenerate declines, likely contributing to sarcopenia, an age-related condition defined as loss of muscle mass and function. Recent studies have shown that resident macrophages in skeletal muscle are highly heterogeneous, and their phenotypes shift during aging, which may exacerbate skeletal muscle deterioration and inefficient regeneration. In this review, we highlight recent insight into the heterogeneity and functional roles of macrophages in skeletal muscle regeneration, particularly as it declines with aging.
Collapse
Affiliation(s)
- Chang-Yi Cui
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
32
|
Nawaz A, Fujisaka S, Kado T, Jeelani I, Tobe K. Heterogeneity of adipose tissue-resident macrophages-beyond M1/M2 paradigm. Diabetol Int 2023; 14:125-133. [PMID: 37090127 PMCID: PMC10113418 DOI: 10.1007/s13340-023-00624-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/17/2023] [Indexed: 04/05/2023]
Abstract
Adipose tissue-resident macrophages (ATMs) are reported to be important for maintaining adipose tissue remodeling and homeostasis. ATMs were classified for the first time in 2007 into the M1 and M2 types. This theory suggests that in the non-obese adipose tissue, the anti-inflammatory, alternatively activated macrophages (AAMs) predominate, and regulate tissue homeostasis, remodeling, and insulin sensitivity. On the other hand, classically activated M1-type macrophages increase rapidly in obesity, secrete inflammatory cytokines, such as TNFα and IL-6, and induce insulin resistance. In recent years, experimental findings that cannot be explained by this theory have been clarified one after another and the theory is being reconsidered. In this review, based on recent findings, we summarize reports on the novel metabolic regulatory functions of ATMs beyond the M1/M2 paradigm.
Collapse
Affiliation(s)
- Allah Nawaz
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama-Shi, Toyama, 930-0194 Japan
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215 USA
| | - Shiho Fujisaka
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama-Shi, Toyama, 930-0194 Japan
| | - Tomonobu Kado
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama-Shi, Toyama, 930-0194 Japan
| | - Ishtiaq Jeelani
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, CA USA
| | - Kazuyuki Tobe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama-Shi, Toyama, 930-0194 Japan
| |
Collapse
|
33
|
Karim M, Iqbal T, Nawaz A, Yaku K, Nakagawa T. Deletion of Nmnat1 in Skeletal Muscle Leads to the Reduction of NAD + Levels but Has No Impact on Skeletal Muscle Morphology and Fiber Types. J Nutr Sci Vitaminol (Tokyo) 2023; 69:184-189. [PMID: 37394423 DOI: 10.3177/jnsv.69.184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a coenzyme that mediates many redox reactions in energy metabolism. NAD+ is also a substrate for ADP-ribosylation and deacetylation by poly (ADP-ribose) polymerase and sirtuin, respectively. Nicotinamide mononucleotide adenylyltransferase 1 (Nmnat1) is a NAD+ biosynthesizing enzyme found in the nucleus. Recent research has shown that the maintaining NAD+ levels is critical for sustaining muscle functions both in physiological and pathological conditions. However, the role of Nmnat1 in skeletal muscle remains unexplored. In this study, we generated skeletal muscle-specific Nmnat1 knockout (M-Nmnat1 KO) mice and investigated its role in skeletal muscle. We found that NAD+ levels were significantly lower in the skeletal muscle of M-Nmnat1 KO mice than in control mice. M-Nmnat1 KO mice, in contrast, had similar body weight and normal muscle histology. Furthermore, the distribution of muscle fiber size and gene expressions of muscle fiber type gene expression were comparable in M-Nmnat1 KO and control mice. Finally, we investigated the role of Nmnat1 in muscle regeneration using cardiotoxin-induced muscle injury model, but muscle regeneration appeared almost normal in M-Nmnat1 KO mice. These findings imply that Nmnat1 has a redundancy in the pathophysiology of skeletal muscle.
Collapse
Affiliation(s)
- Mariam Karim
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama
| | - Tooba Iqbal
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama
| | - Allah Nawaz
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama
| | - Keisuke Yaku
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama
- Research Center for Pre-Disease Science, University of Toyama
| |
Collapse
|