1
|
Chen F, Zhou Y, Du X, Lin S, Wan W, Liu Y, Zou H, Xue Y. In situ sustained and pH-responsive Gas6-loaded microspheres for alleviating acute gouty arthritis by immunoregulation. Colloids Surf B Biointerfaces 2025; 254:114809. [PMID: 40412288 DOI: 10.1016/j.colsurfb.2025.114809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/18/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
Acute gouty arthritis is characterized by sudden, severe attacks of pain, swelling, redness, and tenderness in joints. Managing acute gout attacks requires timely and effective control of the inflammatory reaction caused by monosodium urate (MSU) crystals. In clinical practice, intra-articular drug injection is a common treatment which partially reduces the side effects of oral administration. However, due to the low retention time and persistence of small-molecule drugs in the joints, the effects are often minimal. Therefore, a sustainable and on-demand drug delivery system is warranted. In the present study, a novel in situ sustained and pH-responsive hydrogel microsphere loaded with Gas6 (CMCS@SAG) was fabricated for acute gouty arthritis treatment, and the therapeutic effect was evaluated in vitro and in vivo. The results showed that CMCS@SAG was a spherical porous structure with a diameter of 100 μm. The loaded Gas6 was slowly released and referred to release under acidic pH conditions. In vitro, it was observed that CMCS@SAG effectively inhibited acute inflammatory response induced by macrophages, and enhanced mitochondrial function. In vivo, the joint swelling level decreased, infiltration of inflammation decreased, and multiple immunofluorescences showed significantly increased M2 polarization and anti-inflammatory effects in CMCS@SAG group. These results indicated that CMCS@SAG may be a promising avenue for alleviating acute gouty arthritis.
Collapse
Affiliation(s)
- Fangfang Chen
- Department of Rheumatology, Fujian Medical University Union Hospital, Fuzhou, China; Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
| | - Yingzi Zhou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Xingchen Du
- Department of Rheumatology, First Affiliated Hospital of Soochow University, China
| | - Sang Lin
- Department of Rheumatology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Weiguo Wan
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Yun Liu
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Yu Xue
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Su Y, Huang M, Chen Q, He J, Li S, Wang M. Harnessing β-glucan conjugated quercetin nanocomplex to function as a promising anti-inflammatory agent via macrophage-targeted delivery. Carbohydr Polym 2025; 349:122952. [PMID: 39638531 DOI: 10.1016/j.carbpol.2024.122952] [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: 07/04/2024] [Revised: 10/11/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024]
Abstract
Quercetin, a promising anti-inflammatory agent, faces challenges related to poor bioavailability and limited practical applications. β-glucan, a natural polysaccharide, can be specifically recognized by macrophages, making it an ideal targeting carrier to enhance therapeutic efficacy for macrophage-related dysfunctions. In this study, β-glucan conjugated quercetin nano-complexes (CM-Cur@QT) were developed to target macrophage and alleviate pro-inflammatory response in M1-like macrophages. The results demonstrated that CM-Cur@QT exhibited a spheric shape with an average diameter around 200 nm. FT-IR, 1H NMR, XRD and XPS analyses confirmed the complexation of CM-Cur@QT. This complex showed excellent stability during stimulated digestion, protecting QT from degradation while maintaining favorable antioxidant activity. After complexation, CM-Cur@QT displayed sustained uptake kinetics and enhanced accumulation in macrophages, with a 61.88 % increase compared to individual quercetin after 5 h of incubation. Meanwhile, CM-Cur@QT administration induced evidently cell cycle phases transitions and altered phagocytotic activity in M1-like macrophages. Furthermore, CM-Cur@QT reduced intracellular ROS accumulation, achieving a ROS scavenging rate of up to 49.92 %, compared to 25.59 % in quercetin group. This complex also effectively modulated TNF-a, IL-6 and TGF-β secretion profiles in pro-inflammatory macrophages, outperforming individual QT treatment. Notably, CM-Cur@QT facilitated anti-inflammatory effects while minimizing impacts on inactivated M0 macrophages. These findings underscore the potential of CM-Cur@QT as a promising agent for mitigating inflammatory disorders.
Collapse
Affiliation(s)
- Yuting Su
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China
| | - Manting Huang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China
| | - Qiaochun Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China
| | - Jiayi He
- Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China; College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Siqian Li
- Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China; College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Mingfu Wang
- Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China; College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
3
|
Sanches BDA, Rocha LC, Neto JP, Beguelini MR, Ciena AP, Carvalho HF. Telocytes of the male reproductive system: dynamic tissue organizers. Front Cell Dev Biol 2024; 12:1444156. [PMID: 39469114 PMCID: PMC11513265 DOI: 10.3389/fcell.2024.1444156] [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: 06/05/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
Telocytes are CD34+ interstitial cells that have long cytoplasmic projections (called telopodes), and have been detected in several organs, including those of the male reproductive system. In this brief review we evaluate the role of telocytes in tissue organization of the different organs of the male reproductive system in which these cells were studied. In general terms, telocytes act in the tissue organization through networks of telopodes that separate the epithelia from the stroma, as well as dividing the stroma into different compartments. In addition to this contribution to the structural integrity, there is direct and indirect evidence that such "walls" formed by telocytes also compartmentalize paracrine factors that they or other cells produce, which have a direct impact on morphogenesis and the maintenance of organ cell differentiation, as well as on their normal physiology. Moreover, alterations in telocytes and telopode networks are correlated with pathological conditions in the male reproductive system, in response to profound changes in structural organization of the organs, in inflammation, hyperplasia and cancer. Further studies are necessary to evaluate the molecular pathways telocytes employ in different contexts of physiology and disease.
Collapse
Affiliation(s)
- Bruno D. A. Sanches
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Lara C. Rocha
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), Rio Claro, Brazil
| | - J. Pimentel Neto
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), Rio Claro, Brazil
| | | | - Adriano P. Ciena
- Center of Biological and Health Science, Federal University of Western Bahia (UFOB), Barreiras, Brazil
| | - Hernandes F. Carvalho
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
4
|
Soliman SA. Immunohistochemical-properties of the dermal embryonic telocytes. Sci Rep 2024; 14:13899. [PMID: 38886354 PMCID: PMC11183069 DOI: 10.1038/s41598-024-63802-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
The current investigation aims to study the embryonic dermis formed in the early stages of development and identify the initial interstitial components of the dermis that serve as biological and structural scaffolds for the development of the dermal tissue. To investigate the dermal structure, the current study used morphological and immunological techniques. TCs identified by TEM. They had a cell body and unique podomeres and podoms. They formed a 3D network spread throughout the dermis. Homocellular contact established between them, as well as heterocellular contacts with other cells. Immunohistochemical techniques using specific markers for TCss CD34, CD117, and VEGF confirmed TC identification. TCs represent the major interstitial component in the dermal tissue. They established a 3D network, enclosing other cells and structures. Expression of VEGF by TC promotes angiogenesis. TCs establish cellular contact with sprouting endothelial cells. At the site of cell junction with TCs, cytoskeletal filaments identified and observed to form the pseudopodium core that projects from endothelial cells. TCs had proteolytic properties that expressed MMP-9, CD68, and CD21. Proteolytic activity aids in the removal of components of the extracellular matrix and the phagocytosis of degraded remnants to create spaces to facilitate the development of new dermal structures. In conclusion, TCs organized the scaffold for the development of future dermal structures, including fibrous components and skin appendages. Studying dermal TCs would be interested in the possibility of developing therapeutic strategies for treating different skin disorders and diseases.
Collapse
Affiliation(s)
- Soha A Soliman
- Department of Histology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt.
| |
Collapse
|
5
|
Sanches BDA, Teófilo FBS, Brunet MY, Villapun VM, Man K, Rocha LC, Neto JP, Matsumoto MR, Maldarine JS, Ciena AP, Cox SC, Carvalho HF. Telocytes: current methods of research, challenges and future perspectives. Cell Tissue Res 2024; 396:141-155. [PMID: 38539007 DOI: 10.1007/s00441-024-03888-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/12/2024] [Indexed: 04/28/2024]
Abstract
Telocytes (TCs) are CD34-positive interstitial cells that have long cytoplasmic projections, called telopodes; they have been identified in several organs and in various species. These cells establish a complex communication network between different stromal and epithelial cell types, and there is growing evidence that they play a key role in physiology and pathology. In many tissues, TC network impairment has been implicated in the onset and progression of pathological conditions, which makes the study of TCs of great interest for the development of novel therapies. In this review, we summarise the main methods involved in the characterisation of these cells as well as their inherent difficulties and then discuss the functional assays that are used to uncover the role of TCs in normal and pathological conditions, from the most traditional to the most recent. Furthermore, we provide future perspectives in the study of TCs, especially regarding the establishment of more precise markers, commercial lineages and means for drug delivery and genetic editing that directly target TCs.
Collapse
Affiliation(s)
- Bruno D A Sanches
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil
| | - Francisco B S Teófilo
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil
| | - Mathieu Y Brunet
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Victor M Villapun
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Kenny Man
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, The Netherlands
| | - Lara C Rocha
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), 1515 24 A Ave., Rio Claro, São Paulo, Brazil
| | - Jurandyr Pimentel Neto
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), 1515 24 A Ave., Rio Claro, São Paulo, Brazil
| | - Marta R Matsumoto
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil
| | - Juliana S Maldarine
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil
| | - Adriano P Ciena
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), 1515 24 A Ave., Rio Claro, São Paulo, Brazil
| | - Sophie C Cox
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Hernandes F Carvalho
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Carl von Linnaeus St. Bldg G1, Bertrand Russel Ave., Campinas, São Paulo, Brazil.
| |
Collapse
|
6
|
Wei X, Li H, Chen T, Yang X. Histological study of telocytes in mice intrauterine adhesion model and their positive effect on mesenchymal stem cells in vitro. Cell Biol Int 2024; 48:647-664. [PMID: 38353345 DOI: 10.1002/cbin.12137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/31/2023] [Accepted: 01/28/2024] [Indexed: 04/16/2024]
Abstract
Intrauterine adhesions (IUA), the main cause of secondary infertility in women, result from irreversible fibrotic repair of the endometrium due to inflammation or human factors, accompanied by disruptions in the repair function of endometrial stem cells. This significantly impacts the physical and mental health of women in their childbearing years. Telocytes (TCs), a distinctive type of interstitial cells found in various tissues and organs, play diverse repair functions due to their unique spatial structure. In this study, we conduct the inaugural exploration of the changes in TCs in IUA disease and their potential impact on the function of stem cells. Our results show that in vivo, through double immunofluorescence staining (CD34+/Vimentin+; CD34+/CD31-), as endometrial fibrosis deepens, the number of TCs gradually decreases, telopodes shorten, and the three-dimensional structure becomes disrupted in the mouse IUA mode. In vitro, TCs can promote the proliferation and cycle of bone mesenchymal stem cells (BMSCs) by promoting the Wnt/β-catenin signaling pathway, which were inhibited using XAV939. TCs can promote the migrated ability of BMSCs and contribute to the repair of stem cells during endometrial injury. In addition, TCs can inhibit the apoptosis of BMSCs through the Bcl-2/Bax pathway. In conclusion, our study demonstrates, for the first time, the resistance role of TCs in IUA disease, shedding light on their potential involvement in endometrial repair through the modulation of stem cell function.
Collapse
Affiliation(s)
- Xiaojiao Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, People's Republic of China
| | - Hui Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, People's Republic of China
| | - Tianquan Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, People's Republic of China
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Yangzhou University, Yangzhou City, Jiangsu Province, People's Republic of China
| | - Xiaojun Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, People's Republic of China
| |
Collapse
|
7
|
Wei XJ, Huang YL, Chen TQ, Yang XJ. Inhibitory effect of telocyte-induced M1 macrophages on endometriosis: Targeting angiogenesis and invasion. Acta Histochem 2023; 125:152099. [PMID: 37813067 DOI: 10.1016/j.acthis.2023.152099] [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: 11/29/2022] [Revised: 09/28/2023] [Accepted: 09/30/2023] [Indexed: 10/11/2023]
Abstract
PURPOSE Telocytes (TCs), a novel type of stromal cells found in tissues, induce macrophage differentiation into classically activated macrophages (M1) types and enhance their phagocytic function. The purpose of this study was to investigate the inhibitory effects of TC-induced M1 macrophages on endometriosis (EMs). METHODS mouse uterine primary TCs and endometrial stromal cells (ESCs) were isolated and identified using double immunofluorescence staining. For the in vitro study, ESCs were treated with TC-induced M1 macrophages, and the vascular endothelial growth factor (VEGF), matrix metalloproteinase 9 (MMP9), and nuclear factor kappa B (NF-κb) genes were identified by quantitative real-time PCR (qRT-PCR) or western blotting (WB). For the in vivo study, an EMs mouse model received TC-conditioned medium (TCM) via abdominal administration, and characterized the inhibitory effects on growth (lesion weight, volume, and pathology), tissue-resident macrophages differentiation by immunostaining, angiogenic capacity (CD31 and VEGF), invasive capacity (MMP9), and NF-κb expression within EMs lesions. RESULTS immunofluorescent staining showed that uterine TCs expressed CD34+ and vimentin+, whereas ESCs expressed vimentin+ and cytokeratin-. At the cellular level, TC-induced M1 macrophages can significantly inhibit the expression of VEGF and MMP9 in ESCs through WB or qRT-PCR, possibly by suppressing the NF-κb pathway. The in vivo study showed that macrophages switch from the alternatively activated macrophages (M2) in untreated EMs lesions to the M1 subtype after TCM exposure. Thereby, TC-induced M1 macrophages contributed to the inhibition of EMs lesions. More importantly, this effect may be achieved by suppressing the expression of NF-κb to inhibit angiogenesis (CD31 and VEGF) and invasion (MMP9) in the tissue. CONCLUSION TC-induced M1 macrophages play a prevailing role in suppressing EMs by inhibiting angiogenic and invasive capacity through the NF-κb pathway, which provides a promising therapeutic approach for EMs.
Collapse
Affiliation(s)
- Xiao-Jiao Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province 215006, PR China
| | - Yue-Lin Huang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guiyang City, Guizhou Province 550000, PR China
| | - Tian-Quan Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province 215006, PR China; Department of Obstetrics and Gynecology, The Affiliated Hospital of Yangzhou University, Yangzhou City, Jiangsu Province 225000, PR China
| | - Xiao-Jun Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province 215006, PR China.
| |
Collapse
|
8
|
Gao X, Gao H, Shao W, Wang J, Li M, Liu S. The Extracellular Vesicle-Macrophage Regulatory Axis: A Novel Pathogenesis for Endometriosis. Biomolecules 2023; 13:1376. [PMID: 37759776 PMCID: PMC10527545 DOI: 10.3390/biom13091376] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Endometriosis (EMs) is a common disease among women whose pathogenesis is still unclear, although there are various hypotheses. Recent studies have considered macrophages the key part of the immune system in developing EMs, inducing inflammation, the growth and invasion of the ectopic endometrium, and angiogenesis. Extracellular vesicles (EVs) as novel intercellular vesicle traffic, can be secreted by many kinds of cells, including macrophages. By carrying long non-coding RNA (lncRNA), microRNA (miRNA), or other molecules, EVs can regulate the biological functions of macrophages in an autocrine and paracrine manner, including ectopic lesion growth, immune dysfunction, angiogenesis, and can further accelerate the progression of EMs. In this review, the interactions between macrophages and EVs for the pathogenesis of EMs are summarized. Notably, the regulatory pathways and molecular mechanisms of EVs secreted by macrophages during EMs are reviewed.
Collapse
Affiliation(s)
- Xiaoxiao Gao
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai 201508, China; (X.G.); (H.G.); (W.S.); (J.W.)
| | - Han Gao
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai 201508, China; (X.G.); (H.G.); (W.S.); (J.W.)
| | - Wei Shao
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai 201508, China; (X.G.); (H.G.); (W.S.); (J.W.)
| | - Jiaqi Wang
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai 201508, China; (X.G.); (H.G.); (W.S.); (J.W.)
| | - Mingqing Li
- Laboratory for Reproductive Immunology, Institute Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China
| | - Songping Liu
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai 201508, China; (X.G.); (H.G.); (W.S.); (J.W.)
| |
Collapse
|
9
|
Tian C, Li Y, Si J, Kang J, Chen Z, Nuermaimaiti R, Wang Y, Yu Y, Zhao Z, Wang X, Zhang Y, Zhao H, Yang H, You MJ, Zheng G, Wang L. Knockdown of let-7b in leukemia associated macrophages inhibit acute myeloid leukemia progression. Hematol Oncol 2023; 41:510-519. [PMID: 36579468 DOI: 10.1002/hon.3120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/15/2022] [Accepted: 10/22/2022] [Indexed: 12/30/2022]
Abstract
Macrophages, critical components of bone marrow microenvironment, are reported to be remodeled into leukemia-associated macrophages (LAMs) in leukemic microenvironment where they contribute to leukemia development, characterized as M2 macrophages with pro-tumor effects. However, how leukemic microenvironment transforms macrophages into LAMs remains unknown. Here, we analyzed the clinical relevance of LAMs and profiled their RNA-Seq from acute myeloid leukemia (AML) patients with complete remission (CR) after induction treatment and refractory AML patients. Our results showed that the proportion and number of LAMs in refractory AML patients was higher than that in CR patients and LAM was a poor prognostic factor of AML patients. Furthermore, let-7b was a potentially aberrant gene in LAMs contributed to M2-subtype characteristics. Knockdown of let-7b in LAMs could inhibit the development of AML by repolarizing LAMs toward M1-subtype characteristics through the activation of Toll-like receptor and NF-κB pathway. Our study provides insight for future LAM-based immunotherapy strategies for AML.
Collapse
Affiliation(s)
- Chen Tian
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Hotan District People's Hospital, Hotan, Xinjiang, China
| | - Yueyang Li
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Junqi Si
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Junnan Kang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Zehui Chen
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | | | - Yafei Wang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yong Yu
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhigang Zhao
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiaofang Wang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yizhuo Zhang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Haifeng Zhao
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Hongliang Yang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Mingjian James You
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Guoguang Zheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Lina Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
10
|
Li X, Li X, Yang J, Du Y, Chen L, Zhao G, Ye T, Zhu Y, Xu X, Deng L, Cui W. In Situ Sustained Macrophage-Targeted Nanomicelle-Hydrogel Microspheres for Inhibiting Osteoarthritis. RESEARCH (WASHINGTON, D.C.) 2023; 6:0131. [PMID: 37223475 PMCID: PMC10202383 DOI: 10.34133/research.0131] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 04/10/2023] [Indexed: 05/25/2023]
Abstract
There are still challenges in applying drug nanocarriers for in situ sustained macrophage targeting and regulation, due to the rapid clearance of nanocarriers and burst drug release in vivo. Herein, a nanomicelle-hydrogel microsphere, characterized by its macrophage-targeted nanosized secondary structure that allows it to accurately bind to M1 macrophages through active endocytosis, is employed for in situ sustained macrophage targeting and regulation, and addresses the insufficient osteoarthritis therapeutic efficacy caused by rapid clearance of drug nanocarriers. The 3-dimensional structure of a microsphere can prevent the rapid escape and clearance of a nanomicelle, thus keeping it in joints, while the ligand-guided secondary structure can carry drugs to accurately target and enter M1 macrophages, and release drugs via the transition from hydrophobicity to hydrophilicity of nanomicelles under inflammatory stimulation inside the macrophages. The experiments show that the nanomicelle-hydrogel microsphere can in situ sustainably target and regulate M1 macrophages for more than 14 days in joints, and attenuate local "cytokine storm" by continuous M1 macrophage apoptosis promotion and polarization inhibition. This micro/nano-hydrogel system shows excellent ability to sustainably target and regulate macrophage, realizes the improvement of drug utilization and efficacy inside the macrophage, and thereby can be a potential platform for treating macrophage-related diseases.
Collapse
Affiliation(s)
| | | | | | - Yawei Du
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Liang Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Gang Zhao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Tingjun Ye
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Yuan Zhu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Xiangyang Xu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| |
Collapse
|
11
|
Dama G, Hu X, Yan Y, Li Y, Li H, Yang F, Liu Y, Lin J. Identification and protective role of CD34 + stromal cells/telocytes in experimental autoimmune encephalomyelitis (EAE) mouse spleen. Histochem Cell Biol 2023:10.1007/s00418-023-02186-5. [PMID: 37014442 DOI: 10.1007/s00418-023-02186-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 04/05/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a classical animal model of human multiple sclerosis (MS) that is most commonly used to study the neuropathology and therapeutic effects of the disease. Telocytes (TCs) are a specialized type of interstitial or mesenchymal cell first identified by Popescu in various tissues and organs. However, the existence, distribution and role of CD34+ stromal cells (SCs)/TCs in the EAE-induced mouse spleen remain to be elucidated. We conducted immunohistochemistry, immunofluorescence (double staining for CD34 and c-kit, vimentin, F4/80, CD163, Nanog, Sca-1, CD31 or tryptase) and transmission electron microscopy experiments to investigate the existence, distribution and role of CD34+ SCs/TCs in the EAE-induced mouse spleen. Interestingly, immunohistochemistry, double-immunofluorescence, and transmission electron microscopy results revealed that CD34+ SCs/TCs were significantly upregulated in the EAE mouse spleen. Immunohistochemical or double-immunofluorescence staining of CD34+ SCs/TCs showed positive expression for CD34, c-kit, vimentin, CD34/vimentin, c-kit/vimentin and CD34/c-kit, and negative expression for CD31 and tryptase. Transmission electron microscopy (TEM) results demonstrated that CD34+ SCs/TCs established close connections with lymphocytes, reticular cells, macrophages, endothelial cells and erythrocytes. Furthermore, we also found that M1 (F4/80) or M2 (CD163) macrophages, and haematopoietic, pluripotent stem cells were markedly increased in EAE mice. Our results suggest that CD34+ SCs/TCs are abundant and may play a contributing role in modulating the immune response, recruiting macrophages and proliferation of haematopoietic and pluripotent stem cells following injury to promote tissue repair and regeneration in EAE mouse spleens. This suggests that their transplantation combined with stem cells might represent a promising therapeutic target for the treatment and prevention of multiple autoimmune and chronic inflammatory disorders.
Collapse
Affiliation(s)
- Ganesh Dama
- Henan Joint International Research Laboratory of Stem Cell Medicine, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road #601, Xinxiang City, 453003, Henan Province, China
- Department of Community Health, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200, Kepala Batas, Pulau Pinang, Malaysia
| | - Xiaoxi Hu
- College of Life Sciences and Technology, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road # 601, Xinxiang, 453003, China
| | - Yushan Yan
- College of Life Sciences and Technology, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road # 601, Xinxiang, 453003, China
| | - Yonghai Li
- College of Life Sciences and Technology, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road # 601, Xinxiang, 453003, China
| | - Han Li
- College of Life Sciences and Technology, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road # 601, Xinxiang, 453003, China
| | - Fen Yang
- Henan Joint International Research Laboratory of Stem Cell Medicine, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road #601, Xinxiang City, 453003, Henan Province, China
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yanli Liu
- Henan Joint International Research Laboratory of Stem Cell Medicine, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road #601, Xinxiang City, 453003, Henan Province, China.
- College of Life Sciences and Technology, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road # 601, Xinxiang, 453003, China.
| | - Juntang Lin
- Henan Joint International Research Laboratory of Stem Cell Medicine, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road #601, Xinxiang City, 453003, Henan Province, China.
- College of Life Sciences and Technology, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, East of JinSui Road # 601, Xinxiang, 453003, China.
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
12
|
Zhang X, Sun D, Zhou X, Zhang C, Yin Q, Chen L, Tang Y, Liu Y, Morozova-Roche LA. Proinflammatory S100A9 stimulates TLR4/NF-κB signaling pathways causing enhanced phagocytic capacity of microglial cells. Immunol Lett 2023; 255:54-61. [PMID: 36870421 DOI: 10.1016/j.imlet.2023.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 02/26/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023]
Abstract
Alzheimer's disease (AD) is the main cause of dementia, affecting the increasingly aging population. Growing evidence indicates that neuro-inflammation plays crucial roles, e.g., the association between AD risk genes with innate immune functions. In this study, we demonstrate that moderate concentrations of pro-inflammatory cytokine S100A9 regulate immune response of BV2 microglial cells, i.e., the phagocytic capacity, reflected by elevated number of 1 μm diameter Dsred-stained latex beads in the cytoplasm. In contrast, at high S100A9 concentrations, both the viability and phagocytic capacity of BV2 cells drop substantially. Furthermore, it is uncovered that S100A9 affects phagocytosis of microglia via NF-κB signaling pathways. Application of related target-specific drugs, i.e., IKK and TLR4 inhibitors, effectively suppresses BV2 cells' immune responses. These results suggest that pro-inflammatory S100A9 activates microglial phagocytosis, and possibly contributes to the clearance of amyloidogenic species at the early stage of AD.
Collapse
Affiliation(s)
- Xiaoyin Zhang
- Laboratory of stem cell and Tissue Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Dan Sun
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon Technology, Northwest University, 710127, Xi'an, China
| | - Xin Zhou
- Laboratory of stem cell and Tissue Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Ce Zhang
- State Key Laboratory of Photon-Technology in Western China Energy, Institute of Photonics and Photon Technology, Northwest University, 710127, Xi'an, China
| | - Qing Yin
- Laboratory of stem cell and Tissue Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Li Chen
- Laboratory of stem cell and Tissue Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Yong Tang
- Laboratory of stem cell and Tissue Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Yonggang Liu
- Laboratory of stem cell and Tissue Engineering, Chongqing Medical University, 400016, Chongqing, China.
| | | |
Collapse
|
13
|
Hussein MM, Sayed RKA, Mokhtar DM. Structural and immunohistochemical analysis of the cellular compositions of the liver of molly fish (Poecilia sphenops), focusing on its immune role. ZOOLOGICAL LETTERS 2023; 9:1. [PMID: 36604695 PMCID: PMC9814241 DOI: 10.1186/s40851-022-00200-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 11/21/2022] [Indexed: 06/17/2023]
Abstract
The liver of fish is considered an ideal model for studying the collaboration between environmental agents and the health state of the fish, where it gives good indications about aquatic ecosystem status. Therefore, this study presented immune roles for the liver in molly fish (Poecilia sphenops), using immunohistochemistry and transmission electron microscopy (TEM). The hepatocytes' sinusoidal structures of molly fish livers had taken two different forms; cord-like and tubular, while the biliary tract system showed two different types: isolated and biliary venous tract. The TEM showed that the hepatocytes possessed well-developed cytoplasmic organelles and numerous glycogen and lipid droplets of different sizes. Kupffer cells, Ito cells, aggregation of intrahepatic macrophages and melanomacrophages were also recognized. Melanomacrophages contained numerous phagosomes, many lysosomes, cytoplasmic vacuoles, and melanin pigments. Hepatocytes and Kupffer cells expressed immunoreactivity to APG5, indicating that these cells were involved in the process of autophagy. Telocytes (TCs) were also recognized in the liver of molly fish, and they shared the same morphological characteristics as those in mammals. However, TCs expressed strong immunoreactivity to APG5, TGF-β, and Nrf2, suggesting their possible role in cellular differentiation and regeneration, in addition to phagocytosis and autophagy. Both IL-1β and NF-KB showed immunoreactivity in the hepatocytes and in inflammatory cells (including intrahepatic macrophages and melanomacrophage center). Nrf2 and SOX9 showed immunoreactivity in hepatocytes, stem cells, and macrophages. The present study showed the spatial distribution of hepatic vascular-biliary tracts in molly fish. The liver of molly fish has unique functions in phagocytosis, autophagy, and cell regeneration. The expression of APG5 in hepatocytes, Kupffer cells, melanomacrophages, and telocytes supports the role of the liver in lymphocyte development and proliferation. The expression of TGF-β and NF-κB in hepatocytes, Kupffer cells, telocytes, and macrophages suggests the role of the liver in regulation of cell proliferation and immune response suppression. The expression of IL-1β and Sox9 in macrophages and melanomacrophages suggests the role of the liver in regulation of both innate and adaptive immunity, cell proliferation and apoptosis, in addition to stem cell maintenance.
Collapse
Affiliation(s)
- Marwa M Hussein
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Ramy K A Sayed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, 82524, Sohag, Egypt.
| | - Doaa M Mokhtar
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
- Department of Histology and Anatomy, Faculty of Veterinary Medicine, Badr University in Assiut, Assiut, Egypt
| |
Collapse
|
14
|
Xu T, Zhang H, Zhu Z. Telocytes and endometriosis. Arch Gynecol Obstet 2023; 307:39-49. [PMID: 35668319 DOI: 10.1007/s00404-022-06634-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/14/2022] [Indexed: 02/02/2023]
Abstract
Endometriosis involving the presence and growth of glands and stroma outside the uterine cavity is a common, inflammatory, benign gynecologic disease. Nevertheless, no single theory can exactly account for the pathogenesis of endometriosis. Telocytes, a kind of novel mesenchymal cells, have been suggested to be crucial in promoting angiogenesis and increasing the activity of endometrial interstitial cells and inflammatory cells. Given above roles, telocytes may be considered as the possible pathogenesis of endometriosis. We reviewed the current literature on telocytes. The following aspects were considered: (A) the telocytes' typical characteristics, function, and morphological changes in endometriosis; (B) the potential role of telocytes in endometriosis by impacting the inflammation, invasion, and angiogenesis; (C) telocytes as the potential treatment options for endometriosis.
Collapse
Affiliation(s)
- Ting Xu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No.128, Shenyang Road, Shanghai, 200090, China
| | - Hongqi Zhang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Zhiling Zhu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No.128, Shenyang Road, Shanghai, 200090, China.
| |
Collapse
|
15
|
Chen TQ, Wei XJ, Liu HY, Zhan SH, Yang XJ. Telocyte-Derived Exosomes Provide an Important Source of Wnts That Inhibits Fibrosis and Supports Regeneration and Repair of Endometrium. Cell Transplant 2023; 32:9636897231212746. [PMID: 38006220 PMCID: PMC10676634 DOI: 10.1177/09636897231212746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 09/22/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Intrauterine adhesions (IUAs) often occurred after common obstetrical and gynecological procedures or infections in women of reproductive age. It was characterized by the formation of endometrial fibrosis and prevention of endometrial regeneration, usually with devastating fertility consequences and poor treatment outcomes so far. Telocytes (TCs), as a novel interstitial cell type, present in female uterus with in vitro therapeutic potential in decidualization-defective gynecologic diseases. This study aims to further investigate the role of TC-derived Wnt ligands carried by exosomes (Exo) in reversal of fibrosis and enhancement of regeneration repair in endometrium. IUA cellular and animal models were established from endometrial stromal cells (ESCs) and mice, followed with treatment of TC-conditioned medium (TCM) or TC-derived Exo. In cellular model, fibrosis markers (collagen type 1 alpha 1 [COL1A1], fibronectin [FN], and α-smooth muscle actin [α-SMA]), angiogenesis (vascular endothelial growth factor [VEGF]), and pathway protein (β-catenin) were determined by quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blotting (WB), and immunofluorescence. Results showed that, TCs (either TCM or TC-derived Exo) provide a source of Wnts that inhibit cellular fibrosis, as evidenced by significantly elevated VEGF and β-catenin with decreased fibrotic markers, whereas TCs lost salvage on fibrosis after being blocked with Wnt/β-catenin inhibitors (XAV939 or ETC-159). Further in mouse model, regeneration repair (endometrial thickness, number of glands, and fibrosis area ratio), fibrosis markers (fibronectin [FN]), mesenchymal-epithelial transition (MET) (E-cadherin, N-cadherin), and angiogenesis (VEGF, microvessel density [MVD]) were studied by hematoxylin-eosin (HE), Masson staining, and immunohistochemistry. Results demonstrated that TC-Exo treatment effectively promotes regeneration repair of endometrium by relieving fibrosis, enhancing MET, and angiogenesis. These results confirmed new evidence for therapeutic perspective of TC-derived Exo in IUAs.
Collapse
Affiliation(s)
- Tian-Quan Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xiao-Jiao Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hai-Yan Liu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Sheng-Hua Zhan
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiao-Jun Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
16
|
Jiang Y, Chai X, Chen S, Chen Z, Tian H, Liu M, Wu X. Exosomes from the Uterine Cavity Mediate Immune Dysregulation via Inhibiting the JNK Signal Pathway in Endometriosis. Biomedicines 2022; 10:biomedicines10123110. [PMID: 36551866 PMCID: PMC9775046 DOI: 10.3390/biomedicines10123110] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
Endometriosis is a chronic inflammatory disease with an uncertain pathogenesis. Peritoneal immune dysregulation plays an important role in the pathogenesis of endometriosis. Exosomes are messengers of intercellular communication. This study mainly investigated the role of exosomes from the uterine cavity in endometriosis. Exosomes of the uterine aspirate fluid were isolated and cocultured with macrophages for 48 h. Flow cytometry was used to detect macrophage polarization. A Human MAPK Phosphorylation Antibody Array and Western blot were used to detect the phosphorylation of the MAPK pathway. A microRNA sequencing analysis was used to detect differentially expressed miRNAs. Our research found that exosomes of the uterine aspirate fluid from endometriosis could reduce the proportion of CD80+ macrophages. Additionally, it could inhibit the expression of P-JNK in macrophages. However, the JNK activator anisomycin could increase the proportion of CD80+ macrophages. In addition, exosomes of the uterine aspirate fluid from endometriosis could promote the migration and invasion of endometrial stromal cells by acting on macrophages. The expression of miR-210-3p was increased in both exosomes and the eutopic endometrium in patients with endometriosis through miRNA sequencing, which could also reduce the proportion of CD80+ macrophages. In summary, we propose that exosomes from the uterine cavity in patients with endometriosis may affect the phenotype of macrophages by inhibiting the JNK signaling pathway, thus mediating the formation of an immunological microenvironment conducive to the development of endometriosis.
Collapse
|
17
|
Liu Y, Wang J, Zhang X. An Update on the Multifaceted Role of NF-kappaB in Endometriosis. Int J Biol Sci 2022; 18:4400-4413. [PMID: 35864971 PMCID: PMC9295070 DOI: 10.7150/ijbs.72707] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/12/2022] [Indexed: 11/14/2022] Open
Abstract
Endometriosis remains a common but challenging gynecological disease among reproductive-aged women with an unclear pathogenesis and limited therapeutic options. Numerous pieces of evidence suggest that NF-κB signaling, a major regulator of inflammatory responses, is overactive in endometriotic lesions and contributes to the onset, progression, and recurrence of endometriosis. Several factors, such as estrogen, progesterone, oxidative stress, and noncoding RNAs, can regulate NF-κB signaling in endometriosis. In the present review, we discuss the mechanisms by which these factors regulate NF-κB during endometriosis progression and provide an update on the role of NF-κB in affecting endometriotic cells, peritoneal macrophages (PMs) as well as endometriosis-related symptoms, such as pain and infertility. Furthermore, the preclinical drugs for blocking NF-κB signaling in endometriosis are summarized, including plant-derived medicines, NF-κB inhibitors, other known drugs, and the potential anti-NF-κB drugs predicted through the Drug-Gene Interaction Database. The present review discusses most of the studies concerning the multifaceted role of NF-κB signaling in endometriosis and provides a summary of NF-κB-targeted treatment in detail.
Collapse
Affiliation(s)
- Yuanmeng Liu
- Department of Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Xueshi Road, Hangzhou 310006, China
| | - Jianzhang Wang
- Department of Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Xueshi Road, Hangzhou 310006, China
| | - Xinmei Zhang
- Department of Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Xueshi Road, Hangzhou 310006, China.,Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| |
Collapse
|
18
|
Mokhtar DM, Hussein MM, Sayed RKA. Novel Identification and Microscopy of the Intestinal Bulb of Molly Fish ( Poecilia sphenops) with a Focus on Its Role in Immunity. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2022; 28:1-13. [PMID: 35686429 DOI: 10.1017/s1431927622012089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The intestinal bulb is a simple dilatation in the anterior part of the intestine of agastric fish. This study was conducted on 18 adult specimens of molly fish (Poecilia sphenops) and demonstrated the presence of an intestinal bulb. The intestinal epithelium was composed of enterocytes covered with microvilli, many mucous goblet cells, and enteroendocrine cells. Numerous intraepithelial lymphocytes, neutrophils, plasma cells, dendritic cells, stem cells, rodlet cells, and macrophages were identified in the epithelial layer. Interestingly, this study recorded the process of autophagy and formation of autophagosomes, multivesicular bodies, and dense bodies. The intestinal epithelium extended into the intestinal gland that consisted of simple columnar epithelium, mucous cells, stem cells, enteroendocrine cells, and basal cells. These glands opened to the lumen of the bulb and were surrounded by a network of telocytes. Moreover, immunohistochemistry revealed that the intestinal epithelium expressed APG5, myostatin, TGF-β, IL-1β, NF-κB, Nrf2, and SOX9. Leukocytes in the lamina propria-submucosa expressed APG5. The inflammatory cells in the connective tissue showed strong immunoreactivity to myostatin and TGF-β. The smooth muscular layer also expressed myostatin. Both IL-1β and NF-κB showed immunoreactivity in macrophages in the lamina propria-submucosa. Stem cells expressed Sox-9 and telocytes expressed NF-κB and SOX9; while astrocytes in the tunica muscularis expressed GFAP. The high frequency of immune cells in the intestinal bulb suggested an immune role of this organ. This is the first study demonstrating the absence of the stomach and its replacement with an intestinal bulb in molly fish, and consequently, this species could be reclassified as agastric fish according to this study.
Collapse
Affiliation(s)
- Doaa M Mokhtar
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assuit University, Assiut 71526, Egypt
| | - Marwa M Hussein
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assuit University, Assiut 71526, Egypt
| | - Ramy K A Sayed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt
| |
Collapse
|
19
|
Kobayashi H, Imanaka S. Understanding the molecular mechanisms of macrophage polarization and metabolic reprogramming in endometriosis: A narrative review. Reprod Med Biol 2022; 21:e12488. [PMID: 36310658 PMCID: PMC9596393 DOI: 10.1002/rmb2.12488] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/15/2022] [Accepted: 09/29/2022] [Indexed: 12/03/2022] Open
Abstract
Background Endometriosis is an estrogen-dependent disease and causes pelvic pain and infertility. The limits of current pharmacotherapy in women who desire to become pregnant prompt the development of various targeted molecules for more effective treatment. A review article focused on the unique aspect of cellular metabolic reprogramming of endometriotic cells has been reported. The cellular metabolic pathways are reprogrammed to adapt to a variety of environmental stresses (e.g., nutrient starvation or glucose deprivation, hypoxic stress, excessive reactive oxygen species generation, and other environmental factors). This review aims to summarize macrophage polarization and metabolic reprogramming in endometriosis. Methods A literature search was performed between January 2000 and March 2022 in the PubMed and Google Scholar databases using a combination of specific terms. Results Macrophage cellular metabolism has a marked influence on its phenotype and function. Preclinical studies showed that metabolic conversion toward glycolysis or oxidative phosphorylation drives macrophage polarization to M1 or M2 phenotype, respectively. Such cellular metabolic rewiring can offer new therapeutic opportunities. Conclusion A better understanding of metabolic reprogramming biology in endometriosis-associated macrophages is essential in considering novel therapeutic approach for endometriosis. However, there are currently no detailed studies on therapeutic strategies targeting the cellular metabolic properties of endometriosis-associated macrophages.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of GynecologyMs.Clinic MayOneKashihara, NaraJapan
- Department of Obstetrics and GynecologyNara Medical UniversityKashihara, NaraJapan
| | - Shogo Imanaka
- Department of GynecologyMs.Clinic MayOneKashihara, NaraJapan
- Department of Obstetrics and GynecologyNara Medical UniversityKashihara, NaraJapan
| |
Collapse
|
20
|
Wei XJ, Chen TQ, Yang XJ. Telocytes in Fibrosis Diseases: From Current Findings to Future Clinical Perspectives. Cell Transplant 2022; 31:9636897221105252. [PMID: 35748420 PMCID: PMC9235300 DOI: 10.1177/09636897221105252] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2022] [Indexed: 11/17/2022] Open
Abstract
Telocytes (TCs), a distinct type of interstitial (stromal) cells, have been discovered in many organs of human and mammal animals. TCs, which have unique morphological characteristics and abundant paracrine substance, construct a three-dimensional (3D) interstitial network within the stromal compartment by homocellular and heterocellular communications which are important for tissue homeostasis and normal development. Fibrosis-related diseases remain a common but challenging problem in the field of medicine with unclear pathogenesis and limited therapeutic options. Recently, increasing evidences suggest that where TCs are morphologically or numerically destructed, many diseases continuously develop, finally lead to irreversible interstitial fibrosis. It is not difficult to find that TCs are associated with chronic inflammation and fibrosis. This review mainly discusses relationship between TCs and the occurrence of fibrosis in various diseases. We analyzed in detail the potential roles and speculated mechanisms of TCs in onset and progression of systemic fibrosis diseases, as well as providing the most up-to-date research on the current therapeutic roles of TCs and involved related pathways. Only through continuous research and exploration in the future can we uncover its magic veil and provide strategies for treatment of fibrosis-related disease.
Collapse
Affiliation(s)
- Xiao-jiao Wei
- Department of Obstetrics and
Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, P.R.
China
| | - Tian-quan Chen
- Department of Obstetrics and
Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, P.R.
China
| | - Xiao-jun Yang
- Department of Obstetrics and
Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, P.R.
China
| |
Collapse
|
21
|
Yang L, Bi L, Jin L, Wang Y, Li Y, Li Z, He W, Cui H, Miao J, Wang L. Geniposide Ameliorates Liver Fibrosis Through Reducing Oxidative Stress and Inflammatory Respose, Inhibiting Apoptosis and Modulating Overall Metabolism. Front Pharmacol 2021; 12:772635. [PMID: 34899328 PMCID: PMC8651620 DOI: 10.3389/fphar.2021.772635] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/05/2021] [Indexed: 12/15/2022] Open
Abstract
Liver fibrosis is a progressive liver damage condition caused by various factors and may progress toward liver cirrhosis, and even hepatocellular carcinoma. Many studies have found that the disfunction in metabolism could contribute to the development of liver fibrosis. Geniposide, derived from Gardenia jasminoides J. Ellis, has been demonstrated with therapeutic effects on liver fibrosis. However, the exact molecular mechanisms of such liver-protection remain largely unknown. The aim of this study was to explored the effect of geniposide on metabolic regulations in liver fibrosis. We used carbon tetrachloride (CCl4) to construct a mouse model of liver fibrosis and subsequently administered geniposide treatment. Therapeutic effects of geniposide on liver fibrosis were accessed through measuring the levels of hepatic enzymes in serum and the pathological changes in liver. We also investigated the effects of geniposide on inflammatory response, oxidative stress and apoptosis in liver. Furthermore, serum untargeted metabolomics were used to explore the metabolic regulatory mechanisms behind geniposide on liver fibrosis. Our results demonstrated that geniposide could reduce the levels of hepatic enzymes in serum and ameliorate the pathological changes in liver fibrosis mice. Geniposide enhanced the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) and decreased methane dicarboxylic aldehyde (MDA) levels in liver. Geniposide treatment also decreased the levels of interleukin (IL)-6, IL-1β, and tumor necrosis factor-alpha (TNF-a) in liver tissue homogenate. Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay (TUNEL) staining demonstrated that geniposide could reduce the apoptosis of hepatocytes. Geniposide increased the protein expression of B-cell lymphoma-2 (Bcl-2) and downregulated the protein expression of Bcl-2 Associated X (Bax), cleaved-Caspase 3, and cleaved-Caspase 9. Serum untargeted metabolomics analysis demonstrated that geniposide treatment improved the metabolic disorders including glycerophospholipid metabolism, arginine and proline metabolism, and arachidonic acid (AA) metabolism. In conclusion, our study demonstrated the protective effects of geniposide on liver fibrosis. We found that geniposide could treat liver fibrosis by inhibiting oxidative stress, reducing inflammatory response and apoptosis in the liver, and modulating glycerophospholipid, and arginine, proline, and AA metabolism processes.
Collapse
Affiliation(s)
- Lu Yang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Second People’s Hospital, Tianjin, China
| | - Liping Bi
- Tianjin Second People’s Hospital, Tianjin, China
| | - Lulu Jin
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuming Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuting Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zixuan Li
- Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Wenju He
- Tianjin First Central Hospital, Tianjin, China
| | - Huantian Cui
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Jing Miao
- Tianjin Second People’s Hospital, Tianjin, China
| | - Li Wang
- Tianjin Second People’s Hospital, Tianjin, China
| |
Collapse
|
22
|
Scleroderma-like Impairment in the Network of Telocytes/CD34 + Stromal Cells in the Experimental Mouse Model of Bleomycin-Induced Dermal Fibrosis. Int J Mol Sci 2021; 22:ijms222212407. [PMID: 34830288 PMCID: PMC8620338 DOI: 10.3390/ijms222212407] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 12/19/2022] Open
Abstract
Considerable evidence accumulated over the past decade supports that telocytes (TCs)/CD34+ stromal cells represent an exclusive type of interstitial cells identifiable by transmission electron microscopy (TEM) or immunohistochemistry in various organs of the human body, including the skin. By means of their characteristic cellular extensions (telopodes), dermal TCs are arranged in networks intermingled with a multitude of neighboring cells and, hence, they are thought to contribute to skin homeostasis through both intercellular contacts and releasing extracellular vesicles. In this context, fibrotic skin lesions from patients with systemic sclerosis (SSc, scleroderma) appear to be characterized by a disruption of the dermal network of TCs, which has been ascribed to either cell degenerative processes or possible transformation into profibrotic myofibroblasts. In the present study, we utilized the well-established mouse model of bleomycin-induced scleroderma to gain further insights into the TC alterations found in cutaneous fibrosis. CD34 immunofluorescence revealed a severe impairment in the dermal network of TCs/CD34+ stromal cells in bleomycin-treated mice. CD31/CD34 double immunofluorescence confirmed that CD31-/CD34+ TC counts were greatly reduced in the skin of bleomycin-treated mice compared with control mice. Ultrastructural signs of TC injury were detected in the skin of bleomycin-treated mice by TEM. The analyses of skin samples from mice treated with bleomycin for different times by either TEM or double immunostaining and immunoblotting for the CD34/α-SMA antigens collectively suggested that, although a few TCs may transition to α-SMA+ myofibroblasts in the early disease stage, most of these cells rather undergo degeneration, and then are lost. Taken together, our data demonstrate that TC changes in the skin of bleomycin-treated mice mimic very closely those observed in human SSc skin, which makes this experimental model a suitable tool to (i) unravel the pathological mechanisms underlying TC damage and (ii) clarify the possible contribution of the TC loss to the development/progression of dermal fibrosis. In perspective, these findings may have important implications in the field of skin regenerative medicine.
Collapse
|