1
|
Pan K, Lu Y, Cao D, Peng J, Zhang Y, Li X. Long Non-coding RNA SNHG1 Suppresses the Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells by Binding with HMGB1. Biochem Genet 2024; 62:2869-2883. [PMID: 38038773 DOI: 10.1007/s10528-023-10564-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023]
Abstract
Osteoporosis (OP) has a significant detrimental impact on the health of the elder. Long-term clinical effectiveness of current drugs used for OP treatment is limited. Therefore, it is very important to explore novel treatment targets for OP. The expression of SNHG1, HMGB1, OCN and OPN in gene level was measured using RT-qPCR, and the protein expression was determined by Western blotting assay. The concentration of IL-1β and IL-18 in supernatant of the bone marrow mesenchymal stem cells (BMSCs) was measured by ELISA. The interaction between SNHG1 and HMGB1 was confirmed by RNA pull down. Besides, alizarin red staining was performed to evaluate the differentiation of BMSCs into osteoblast. SNHG1 and HMGB1 were found to be upregulated in the serum of OP patients. During the osteogenic differentiation of BMSCs, the expression of osteoblastogenesis markers (OCN and OPN) and the activity of ALP were upregulated, while the expression levels of SNHG1 and HMGB1 were decreased in a time-dependent manner. In addition, the interaction between SNHG1 and HMGB1, expression of pyroptosis-associated factors (caspase-1 p20 and GSDMD-N), and secretion of IL-1β and IL-18 were also decreased during osteogenic differentiation. Interestingly, increasing SNHG1 promoted HMGB1 expression, activated pyroptosis, but inhibited osteogenic differentiation. Silencing HMGB1 or inhibiting caspase-1 partially rescued the inhibitory effect of SNHG1 on osteogenic differentiation. Our findings indicate that SNHG1 suppresses the osteogenic differentiation of BMSCs by activating pyroptosis through interaction with HMGB1 and promotion of HMGB1 expression. Our work provides further evidence supporting SNHG1 acts as a potential target for OP treatment, and reveals for the first time that SNHG1 regulates osteogenic differentiation by affecting pyroptosis.
Collapse
Affiliation(s)
- Kaihua Pan
- Department of Orthopaedics, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410005, Hunan, People's Republic of China
| | - Yuanyuan Lu
- Department of Orthopaedics, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410005, Hunan, People's Republic of China
| | - Daning Cao
- Department of Orthopaedics, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410005, Hunan, People's Republic of China
| | - Jiang Peng
- Department of Orthopaedics, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410005, Hunan, People's Republic of China
| | - Yunqing Zhang
- Department of Orthopaedics, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410005, Hunan, People's Republic of China
| | - Xiaoming Li
- Department of Orthopaedics, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410005, Hunan, People's Republic of China.
| |
Collapse
|
2
|
Lu P, Dai G, Shi L, Li Y, Zhang M, Wang H, Rui Y. HMGB1 Modulates High Glucose-Induced Erroneous Differentiation of Tendon Stem/Progenitor Cells through RAGE/ β-Catenin Pathway. Stem Cells Int 2024; 2024:2335270. [PMID: 38633380 PMCID: PMC11022503 DOI: 10.1155/2024/2335270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/13/2024] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
The association of tendinopathy with diabetes has been well recognized. Tendon stem/progenitor cells (TSPCs) play critical roles in tendon repair, regeneration, and homeostasis maintenance. Diabetic TSPCs exhibit enhanced erroneous differentiation and are involved in the pathogenesis of diabetic tendinopathy, whereas the underlying mechanism of the erroneous differentiation of TSPCs remains unclear. Here, we showed that high glucose treatment promoted the erroneous differentiation of TSPCs with increased osteogenic differentiation capacity and decreased tenogenic differentiation ability, and stimulated the expression and further secretion of HMGB1 in TSPCs and. Functionally, exogenous HMGB1 significantly enhanced the erroneous differentiation of TSPCs, while HMGB1 knockdown mitigated high glucose-promoted erroneous differentiation of TSPCs. Mechanistically, the RAGE/β-catenin signaling was activated in TSPCs under high glucose, and HMGB1 knockdown inhibited the activity of RAGE/β-catenin signaling. Inhibition of RAGE/β-catenin signaling could ameliorate high glucose-induced erroneous differentiation of TSPCs. These results indicated that HMGB1 regulated high glucose-induced erroneous differentiation of TSPCs through the RAGE/β-catenin signaling pathway. Collectively, our findings suggest a novel essential mechanism of the erroneous differentiation of TSPCs, which might contribute to the pathogenesis of diabetic tendinopathy and provide a promising therapeutic target and approach for diabetic tendinopathy.
Collapse
Affiliation(s)
- Panpan Lu
- Department of Orthopaedics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| | - Guangchun Dai
- Department of Orthopaedics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| | - Liu Shi
- Department of Orthopaedics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| | - Yingjuan Li
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Department of Geriatrics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| | - Ming Zhang
- Department of Orthopaedics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| | - Hao Wang
- Department of Orthopaedics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| | - Yunfeng Rui
- Department of Orthopaedics, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- School of Medicine, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Orthopaedic Trauma Institute (OTI), Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
- Trauma Center, Zhongda Hospital, Southeast University, No 87 Ding Jia Qiao, Nanjing 210009, Jiangsu, China
| |
Collapse
|
3
|
Liao M, Cao J, Chen W, Wang M, Jin Z, Ye J, Ren Y, Wei Y, Xue Y, Chen D, Zhang Y, Chen S. HMGB1 prefers to interact with structural RNAs and regulates rRNA methylation modification and translation in HeLa cells. BMC Genomics 2024; 25:345. [PMID: 38580917 PMCID: PMC10996203 DOI: 10.1186/s12864-024-10204-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 03/08/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND High-mobility group B1 (HMGB1) is both a DNA binding nuclear factor modulating transcription and a crucial cytokine that mediates the response to both infectious and noninfectious inflammation such as autoimmunity, cancer, trauma, and ischemia reperfusion injury. HMGB1 has been proposed to control ribosome biogenesis, similar as the other members of a class of HMGB proteins. RESULTS Here, we report that HMGB1 selectively promotes transcription of genes involved in the regulation of transcription, osteoclast differentiation and apoptotic process. Improved RNA immunoprecipitation by UV cross-linking and deep sequencing (iRIP-seq) experiment revealed that HMGB1 selectively bound to mRNAs functioning not only in signal transduction and gene expression, but also in axon guidance, focal adhesion, and extracellular matrix organization. Importantly, HMGB1-bound reads were strongly enriched in specific structured RNAs, including the domain II of 28S rRNA, H/ACA box snoRNAs including snoRNA63 and scaRNAs. RTL-P experiment showed that overexpression of HMGB1 led to a decreased methylation modification of 28S rRNA at position Am2388, Cm2409, and Gm2411. We further showed that HMGB1 overexpression increased ribosome RNA expression levels and enhanced protein synthesis. CONCLUSION Taken together, our results support a model in which HMGB1 binds to multiple RNA species in human cancer cells, which could at least partially contribute to HMGB1-modulated rRNA modification, protein synthesis function of ribosomes, and differential gene expression including rRNA genes. These findings provide additional mechanistic clues to HMGB1 functions in cancers and cell differentiation.
Collapse
Affiliation(s)
- Meimei Liao
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China
| | - Jiarui Cao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China
| | - Wen Chen
- Center for Genome Analysis, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
| | - Mengwei Wang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China
| | - Zhihui Jin
- Department of Orthopedics, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China
| | - Jia Ye
- Department of Orthopedics, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China
| | - Yijun Ren
- Department of Orthopedics, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China
| | - Yaxun Wei
- Center for Genome Analysis, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
| | - Yaqiang Xue
- Center for Genome Analysis, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
| | - Dong Chen
- Center for Genome Analysis, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
| | - Yi Zhang
- Center for Genome Analysis, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
| | - Sen Chen
- Department of Orthopedics, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China.
| |
Collapse
|
4
|
Wang X, Mijiti W, Jia Q, Yi Z, Ma J, Zhou Z, Xie Z. Exploration of altered miRNA expression and function in MSC-derived extracellular vesicles in response to hydatid antigen stimulation. Front Microbiol 2024; 15:1381012. [PMID: 38601938 PMCID: PMC11004373 DOI: 10.3389/fmicb.2024.1381012] [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: 02/02/2024] [Accepted: 03/14/2024] [Indexed: 04/12/2024] Open
Abstract
Background Hydatid disease is caused by Echinococcus parasites and can affect various tissues and organs in the body. The disease is characterized by the presence of hydatid cysts, which contain specific antigens that interact with the host's immune system. Mesenchymal stem cells (MSCs) are pluripotent stem cells that can regulate immunity through the secretion of extracellular vesicles (EVs) containing microRNAs (miRNAs). Methods In this study, hydatid antigens were isolated from sheep livers and mice peritoneal cavities. MSCs derived from mouse bone marrow were treated with different hydatid antigens, and EVs were isolated and characterized from the conditioned medium of MSCs. Small RNA library construction, miRNA target prediction, and differential expression analysis were conducted to identify differentially expressed miRNAs. Functional enrichment and network construction were performed to explore the biological functions of the target genes. Real-time PCR and Western blotting were used for miRNA and gene expression verification, while ELISA assays quantified TNF, IL-1, IL-6, IL-4, and IL-10 levels in cell supernatants. Results The study successfully isolated hydatid antigens and characterized MSC-derived EVs, demonstrating the impact of antigen concentration on MSC viability. Key differentially expressed miRNAs, such as miR-146a and miR-9-5p, were identified, with functional analyses revealing significant pathways like Endocytosis and MAPK signaling associated with these miRNAs' target genes. The miRNA-HUB gene regulatory network identified crucial miRNAs and HUB genes, such as Traf1 and Tnf, indicating roles in immune modulation and osteogenic differentiation. Protein-protein interaction (PPI) network analysis highlighted central HUB genes like Akt1 and Bcl2. ALP activity assays confirmed the influence of antigens on osteogenic differentiation, with reduced ALP activity observed. Expression analysis validated altered miRNA and chemokine expression post-antigen stimulation, with ELISA analysis showing a significant reduction in CXCL1 expression in response to antigen exposure. Conclusion This study provides insights into the role of MSC-derived EVs in regulating parasite immunity. The findings suggest that hydatid antigens can modulate the expression of miRNAs in MSC-derived EVs, leading to changes in chemokine expression and osteogenic capacity. These findings contribute to a better understanding of the immunomodulatory mechanisms involved in hydatid disease and provide potential therapeutic targets for the development of new treatment strategies.
Collapse
Affiliation(s)
- Xin Wang
- Department of Orthopedics and Trauma, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Wubulikasimu Mijiti
- Department of Orthopedics and Trauma, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Qiyu Jia
- Department of Orthopedics and Trauma, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Zhifei Yi
- Department of Orthopedics and Trauma, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Junchao Ma
- Department of Orthopedics and Trauma, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Ziyu Zhou
- Department of Orthopedics and Trauma, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Zengru Xie
- Department of Orthopedics and Trauma, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
- Key Laboratory of High Incidence Disease Research in Xingjiang (Xinjiang Medical University), Ministry of Education, Ürümqi, Xinjiang, China
- Xinjiang Clinical Research Center for Orthopedics, Xinjiang Medical University, Ürümqi, Xinjiang, China
| |
Collapse
|
5
|
Liu W, Ming S, Zhao X, Zhu X, Gong Y. Developmental expression of high-mobility group box 1 (HMGB1) in the mouse cochlea. Eur J Histochem 2023; 67:3704. [PMID: 37667832 PMCID: PMC10518653 DOI: 10.4081/ejh.2023.3704] [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: 03/09/2023] [Accepted: 08/18/2023] [Indexed: 09/06/2023] Open
Abstract
The expression changes of high-mobility group box 1 (HMGB1) in the mouse cochlea have recently been implicated in noise-induced hearing loss, suggesting that HMGB1 participates in regulating cochlear function. However, the precise role of HMGB1 in the auditory system remains largely unclear. This study aimed to investigate its function in the developing mouse cochlea by examining the expression pattern of HMGB1 in the mouse cochlea from embryonic day (E) 18.5 to postnatal day (P) 28 using double immunofluorescence on frozen sections. Our findings revealed that HMGB1 was extensively expressed in the cell nucleus across various regions of the mouse cochlea, including the organ of Corti. Furthermore, its expression underwent developmental regulation during mouse cochlear development. Specifically, HMGB1 was found to be localized in the tympanic border cells at each developmental stage, coinciding with the gradual anatomical in this region during development. In addition, HMGB1 was expressed in the greater epithelial ridge (GER) and supporting cells of the organ of Corti, as validated by the supporting cell marker Sox2 at P1 and P8. However, at P14, the expression of HMGB1 disappeared from the GER, coinciding with the degeneration of the GER into the inner sulcus cells. Moreover, we observed that HMGB1 co-localized with Ki-67-positive proliferating cells in several cochlear regions during late embryonic and early postnatal stages, including the GER, the tympanic border cells, cochlear lateral wall, and cochlear nerves. Furthermore, by dual-staining Ki-67 with neuronal marker TUJ1 and glial marker Sox10, we determined the expression of Ki-67 in the neonatal glial cells. Our spatial-temporal analysis demonstrated that HMGB1 exhibited distinct expression patterns during mouse cochlear development. The co-localization of HMGB1 with Ki-67-positive proliferating cells suggested that HMGB1 may play a role in cochlear development.
Collapse
Affiliation(s)
- Wenjing Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing.
| | - Shanshan Ming
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing.
| | - Xiaobing Zhao
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing.
| | - Xin Zhu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing.
| | - Yuxiang Gong
- Department of Nephrology, Zhongda Hospital, Southeast University, Nanjing.
| |
Collapse
|
6
|
Intraglandular mesenchymal stem cell treatment induces changes in the salivary proteome of irradiated patients. COMMUNICATIONS MEDICINE 2022; 2:160. [PMID: 36496530 PMCID: PMC9735277 DOI: 10.1038/s43856-022-00223-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Hyposalivation and xerostomia (dry mouth), are the leading site-effects to treatment of head and neck cancer. Currently, there are no effective therapies to alleviate radiation-induced hyposalivation. Adipose tissue-derived mesenchymal stem/stromal cells (AT-MSCs) have shown potential for restoring salivary gland function. However, the mode of action is unknown. The purpose of the present study was therefore to characterize the effect of AT-MSC therapy on the salivary proteome in previously irradiated head and neck cancer patients. METHODS Whole saliva was collected from patients with radiation-induced salivary gland hypofunction (n = 8) at baseline, and 120 days after AT-MSC treatment, and from healthy controls (n = 10). The salivary proteome was characterized with mass spectrometry based proteomics, and data was compared within the AT-MSC group (baseline versus day 120) and between AT-MSC group and healthy controls. Significance levels between groups were determined by using double-sided t-test, and visualized by means of principal component analysis, volcano plots and cluster analysis. RESULTS Here we show that 140 human proteins are significantly differentially expressed in saliva from patients with radiation-induced hypofunction versus healthy controls. AT-MSC treatment induce a significant impact on the salivary proteome, as 99 proteins are differentially expressed at baseline vs. 120 days after treatment. However, AT-MSC treatment does not restore healthy conditions, as 212 proteins are significantly differentially expressed in saliva 120 days after AT-MSCs treatment, as compared to healthy controls. CONCLUSION The results indicate an increase in proteins related to tissue regeneration in AT-MSCs treated patients. Our study demonstrates the impact of AT-MSCs on the salivary proteome, thereby providing insight into the potential mode of action of this novel treatment approach.
Collapse
|
7
|
High Mobility Group Box-1 regulates expression of EGFR, VEGF, StAR and TIMP1/2 in bovine granulosa cells through a mechanism involving TLR2/NF-κB. Anim Reprod Sci 2022; 247:107152. [DOI: 10.1016/j.anireprosci.2022.107152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022]
|
8
|
Ma L, Ke W, Liao Z, Feng X, Lei J, Wang K, Wang B, Li G, Luo R, Shi Y, Zhang W, Song Y, Sheng W, Yang C. Small extracellular vesicles with nanomorphology memory promote osteogenesis. Bioact Mater 2022; 17:425-438. [PMID: 35386457 PMCID: PMC8964989 DOI: 10.1016/j.bioactmat.2022.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/02/2021] [Accepted: 01/04/2022] [Indexed: 12/18/2022] Open
Abstract
Nanotopographical cues endow biomaterials the ability to guide cell adhesion, proliferation, and differentiation. Cellular mechanical memory can maintain the cell status by retaining cellular information obtained from past mechanical microenvironments. Here, we propose a new concept “morphology memory of small extracellular vesicles (sEV)” for bone regeneration. We performed nanotopography on titanium plates through alkali and heat (Ti8) treatment to promote human mesenchymal stem cell (hMSC) differentiation. Next, we extracted the sEVs from the hMSC, which were cultured on the nanotopographical Ti plates for 21 days (Ti8-21-sEV). We demonstrated that Ti8-21-sEV had superior pro-osteogenesis ability in vitro and in vivo. RNA sequencing further confirmed that Ti8-21-sEV promote bone regeneration through osteogenic-related pathways, including the PI3K-AKT signaling pathway, MAPK signaling pathway, focal adhesion, and extracellular matrix-receptor interaction. Finally, we decorated the Ti8-21-sEV on a 3D printed porous polyetheretherketone scaffold. The femoral condyle defect model of rabbits was used to demonstrate that Ti8-21-sEV had the best bone ingrowth. In summary, our study demonstrated that the Ti8-21-sEV have memory function by copying the pro-osteogenesis information from the nanotopography. We expect that our study will encourage the discovery of other sEV with morphology memory for tissue regeneration. Nanotopography fabricated on titanium plates has superior promoted hMSCs differentiation ability. sEV extracted from hMSCs which were cultured on Ti8 plates for 21 days had the superior pro-osteogenesis ability. Ti8-21-sEV have memory function through copy the pro-osteogenesis information from nanotopography. RNA sequencing confirmed that Ti8-21-sEV promote bone regeneration through osteogenic-related pathways.
Collapse
|
9
|
Circ_0099630 Participates in SPRY1-Mediated Repression in Periodontitis. Int Dent J 2022; 73:136-143. [PMID: 35933226 PMCID: PMC9875226 DOI: 10.1016/j.identj.2022.06.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Periodontitis is chronic inflammation that causes damage to periodontal tissues and cementum. It has been reported that circular RNA hsa_circ_0099630 (circ_0099630) was overexpressed in gingival samples from patients with periodontitis. However, the function of circ_0099630 on the osteogenic differentiation of periodontal ligament cells (PDLCs) in periodontitis remains unclear. METHODS Periodontal ligaments from patients with periodontitis and third molars (termed wisdom teeth) were utilised to isolate inflamed PDLCs (iPDLCs) and healthy PDLCs (hPDLCs). Expression levels of circ_0099630 in isolated PDLCs were assessed using reverse transcription quantitative polymerase chain reaction (RT-qPCR). Effects of circ_0099630 overexpression and silencing on iPDLC viability, proliferation, and cycle progression were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), 5-ethynyl-2'-deoxyuridine (EdU), and flow cytometry assays. The osteogenic differentiation was detected by analysing the alkaline phosphatase (ALP) activity, mineralisation amount, and osteogenic markers osterix (OSX), ALP, and RUNX2 in iPDLCs. The regulatory mechanism of circ_0099630 was predicted by bioinformatics analysis and validated by dual-luciferase reporter, RNA immunoprecipitation, and RNA pull-down assays. RESULTS Circ_0099630 was underexpressed in iPDLCs compared to hPDLCs. Overexpression of circ_0099630 repressed iPDLC proliferation and osteogenic differentiation, but circ_0099630 silencing exerted an opposing effect. Mechanically, circ_0099630 sponged miR-212-5p to block the inhibiting effect of miR-212-5p on SPRY1. Elevated expression of SPRY1 partly reversed the promoting effect of circ_0099630 knockdown on iPDLC proliferation and osteogenic differentiation. CONCLUSIONS Circ_0099630 curbed PDLC proliferation and osteogenic differentiation through elevating SPRY1 expression via sponging miR-212-5p in periodontitis.
Collapse
|
10
|
Kishi S, Fujiwara-Tani R, Honoki K, Sasaki R, Mori S, Ohmori H, Sasaki T, Miyagawa Y, Kawahara I, Kido A, Tanaka Y, Kuniyasu H. Oxidized high mobility group B-1 enhances metastability of colorectal cancer via modification of mesenchymal stem/stromal cells. Cancer Sci 2022; 113:2904-2915. [PMID: 35570394 PMCID: PMC9357642 DOI: 10.1111/cas.15400] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/28/2022] [Accepted: 05/10/2022] [Indexed: 11/26/2022] Open
Abstract
High mobility group box-1 (HMGB1) is known to be a chemotactic factor for mesenchymal stem/stromal cells (MSCs), but the effect of post-translational modification on its function is not clear. In this study, we hypothesized that differences in the oxidation state of HMGB1 would lead to differences in the function of MSCs in cancer. In human colorectal cancer, MSCs infiltrating into the stroma were correlated with liver metastasis and serum HMGB1. In animal models, oxidized HMGB1 mobilized 3-fold fewer MSCs to subcutaneous tumors compared to reduced HMGB1. Reduced HMGB1 inhibited proliferation of mouse bone marrow MSCs (BM-MSCs) and induced differentiation into osteoblasts and vascular pericytes, whereas oxidized HMGB1 promoted proliferation and increased stemness, and no differentiation was observed. When BM-MSCs pretreated with oxidized HMGB1 were co-cultured with syngeneic cancer cells, cell proliferation and stemness of cancer cells were increased, and tumorigenesis and drug resistance were promoted. In contrast, co-culture with reduced HMGB1-pretreated BM-MSCs did not enhance stemness. In an animal orthotopic transplantation colorectal cancer model, oxidized HMGB1, but not reduced HMGB1, promoted liver metastasis with intratumoral MSC chemotaxis. Thus, oxidized HMGB1 reprograms MSCs and promotes cancer malignancy. The oxidized HMGB1-MSC axis may be an important target for cancer therapy.
Collapse
Affiliation(s)
- Shingo Kishi
- Department of Molecular Pathology, Nara Medical University, Nara, Japan
| | | | - Kanya Honoki
- Department of Orthopedics, Nara Medical University, Nara, Japan
| | - Rika Sasaki
- Department of Molecular Pathology, Nara Medical University, Nara, Japan
| | - Shiori Mori
- Department of Molecular Pathology, Nara Medical University, Nara, Japan
| | - Hitoshi Ohmori
- Department of Molecular Pathology, Nara Medical University, Nara, Japan
| | - Takamitsu Sasaki
- Department of Molecular Pathology, Nara Medical University, Nara, Japan
| | | | - Isao Kawahara
- Department of Molecular Pathology, Nara Medical University, Nara, Japan
| | - Akira Kido
- Department of Orthopedics, Nara Medical University, Nara, Japan
| | - Yasuhito Tanaka
- Department of Orthopedics, Nara Medical University, Nara, Japan
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, Nara, Japan
| |
Collapse
|
11
|
Kim J, Lee KM, Han SH, Ko EA, Yoon DS, Park IK, Shin HC, Park KH, Lee JW. Development of stabilized dual growth factor-loaded hyaluronate collagen dressing matrix. J Tissue Eng 2021; 12:2041731421999750. [PMID: 33796249 PMCID: PMC7970265 DOI: 10.1177/2041731421999750] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 02/13/2021] [Indexed: 12/26/2022] Open
Abstract
Patients with diabetes experience impaired growth factor production such as
epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF), and
they are reportedly involved in wound healing processes. Here, we report dual
growth factor-loaded hyaluronate collagen dressing (Dual-HCD) matrix, using
different ratios of the concentration of stabilized growth
factors—stabilized-EGF (S-EGF) and stabilized-bFGF (S-bFGF). At first, the
optimal concentration ratio of S-EGF to S-bFGF in the Dual-HCD matrix is
determined to be 1:2 in type I diabetic mice. This Dual-HCD matrix does not
cause cytotoxicity and can be used in vivo. The wound-healing effect of this
matrix is confirmed in type II diabetic mice. Dual HCD enhances angiogenesis
which promotes wound healing and thus, it shows a significantly greater
synergistic effect than the HCD matrix loaded with a single growth factor.
Overall, we conclude that the Dual-HCD matrix represents an effective
therapeutic agent for impaired diabetic wound healing.
Collapse
Affiliation(s)
- Jihyun Kim
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyoung-Mi Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung Hwan Han
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul, South Korea
| | - Eun Ae Ko
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Dong Suk Yoon
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Ik Kyu Park
- R&D Center, Genewel Co., Ltd., Sungnam, South Korea
| | - Hang-Cheol Shin
- School of Systems Biomedical Science, Soongsil University, Seoul, South Korea
| | - Kwang Hwan Park
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin Woo Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
12
|
Subcellular localization of HMGB1 in colorectal cancer impacts on tumor grade and survival prognosis. Sci Rep 2020; 10:18587. [PMID: 33122771 PMCID: PMC7596050 DOI: 10.1038/s41598-020-75783-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022] Open
Abstract
The high-mobility group box-1 (HMGB1) protein is implicated in the development of various cancers and their proliferation. According to its function, HMGB1 shuttles between the cell nucleus and cytoplasm, assisting with nucleosome stabilization and gene transcription, or localizing in the cell membrane for outgrowth. The clinicopathologic and prognostic significance of these different subcellular locations and their correlation has been unclear in colorectal cancer (CRC). We found significantly higher rates of nuclear HMGB1 expression in CRC and colorectal adenoma tissue samples (84.0% and 92.6%, respectively) than in normal colorectal tissue (15.0%) and a significantly higher rate of positive cytoplasmic HMGB1 expression in CRC tissue (25.2%) compared with colorectal adenoma (11.8%) and normal colorectal tissue (0.0%). Positive cytoplasmic HMGB1 expression was associated with high-grade CRC, a poor prognosis, and was negatively correlated with strongly positive nuclear HMGB1 expression in CRC tissue specimens (r = – 0.377, P = 0.000). CRC patients with strongly positive nuclear HMGB1 expression had a better survival prognosis than other CRC patients. Preventing nuclear plasma translocation of HMGB1 may be a new strategy for CRC management.
Collapse
|
13
|
Osteocyte apoptosis: the roles and key molecular mechanisms in resorption-related bone diseases. Cell Death Dis 2020; 11:846. [PMID: 33046704 PMCID: PMC7552426 DOI: 10.1038/s41419-020-03059-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 01/18/2023]
Abstract
Vital osteocytes have been well known to function as an important orchestrator in the preservation of robustness and fidelity of the bone remodeling process. Nevertheless, some key pathological factors, such as sex steroid deficiency and excess glucocorticoids, and so on, are implicated in inducing a bulk of apoptotic osteocytes, subsequently resulting in resorption-related bone loss. As much, osteocyte apoptosis, under homeostatic conditions, is in an optimal state of balance tightly controlled by pro- and anti-apoptotic mechanism pathways. Importantly, there exist many essential signaling proteins in the process of osteocyte apoptosis, which has a crucial role in maintaining a homeostatic environment. While increasing in vitro and in vivo studies have established, in part, key signaling pathways and cross-talk mechanism on osteocyte apoptosis, intrinsic and complex mechanism underlying osteocyte apoptosis occurs in various states of pathologies remains ill-defined. In this review, we discuss not only essential pro- and anti-apoptotic signaling pathways and key biomarkers involved in these key mechanisms under different pathological agents, but also the pivotal role of apoptotic osteocytes in osteoclastogenesis-triggered bone loss, hopefully shedding new light on the attractive and proper actions of pharmacotherapeutics of targeting apoptosis and ensuing resorption-related bone diseases such as osteoporosis and fragility fractures.
Collapse
|
14
|
Jauković A, Kukolj T, Obradović H, Okić-Đorđević I, Mojsilović S, Bugarski D. Inflammatory niche: Mesenchymal stromal cell priming by soluble mediators. World J Stem Cells 2020; 12:922-937. [PMID: 33033555 PMCID: PMC7524701 DOI: 10.4252/wjsc.v12.i9.922] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/13/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are adult stem cells of stromal origin that possess self-renewal capacity and the ability to differentiate into multiple mesodermal cell lineages. They play a critical role in tissue homeostasis and wound healing, as well as in regulating the inflammatory microenvironment through interactions with immune cells. Hence, MSCs have garnered great attention as promising candidates for tissue regeneration and cell therapy. Because the inflammatory niche plays a key role in triggering the reparative and immunomodulatory functions of MSCs, priming of MSCs with bioactive molecules has been proposed as a way to foster the therapeutic potential of these cells. In this paper, we review how soluble mediators of the inflammatory niche (cytokines and alarmins) influence the regenerative and immunomodulatory capacity of MSCs, highlighting the major advantages and concerns regarding the therapeutic potential of these inflammatory primed MSCs. The data summarized in this review may provide a significant starting point for future research on priming MSCs and establishing standardized methods for the application of preconditioned MSCs in cell therapy.
Collapse
Affiliation(s)
- Aleksandra Jauković
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11129, Serbia
| | - Tamara Kukolj
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11129, Serbia
| | - Hristina Obradović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11129, Serbia
| | - Ivana Okić-Đorđević
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11129, Serbia
| | - Slavko Mojsilović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11129, Serbia
| | - Diana Bugarski
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11129, Serbia
| |
Collapse
|
15
|
Abdoli Shadbad M, Hajiasgharzadeh K, Baradaran B. Cross-talk between myeloid-derived suppressor cells and Mucin1 in breast cancer vaccination: On the verge of a breakthrough. Life Sci 2020; 258:118128. [PMID: 32710947 DOI: 10.1016/j.lfs.2020.118128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 01/22/2023]
Abstract
Although breast cancer is one of the leading troublesome cancers, the available therapeutic options have not fulfilled the desired outcomes. Immune-based therapy has gained special attention for breast cancer treatment. Although this approach is highly tolerable, its low response rate has rendered it as an undesirable approach. This review aims to describe the essential oncogenic pathways involved in breast cancer, elucidate the immunosuppression and oncogenic effect of Mucin1, and introduce myeloid-derived suppressor cells, which are the main culprits of anti-tumoral immune response attenuation. The various auto-inductive loops between Mucin1 and myeloid-derived suppressor cells are focal in the suppression of anti-tumoral immune responses in patients with breast cancer. These cross-talks between the Mucin1 and myeloid-derived suppressor cells can be the underlying causes of immunotherapy's impotence for patients with breast cancer. This approach can pave the road for the development of a potent vaccine for patients with breast cancer and is translated into clinical settings.
Collapse
Affiliation(s)
| | - Khalil Hajiasgharzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
Yuan S, Liu Z, Xu Z, Liu J, Zhang J. High mobility group box 1 (HMGB1): a pivotal regulator of hematopoietic malignancies. J Hematol Oncol 2020; 13:91. [PMID: 32660524 PMCID: PMC7359022 DOI: 10.1186/s13045-020-00920-3] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a nonhistone chromatin-associated protein that has been widely reported to play a pivotal role in the pathogenesis of hematopoietic malignancies. As a representative damage-associated molecular pattern (DAMP), HMGB1 normally exists inside cells but can be secreted into the extracellular environment through passive or active release. Extracellular HMGB1 binds with several different receptors and interactors to mediate the proliferation, differentiation, mobilization, and senescence of hematopoietic stem cells (HSCs). HMGB1 is also involved in the formation of the inflammatory bone marrow (BM) microenvironment by activating proinflammatory signaling pathways. Moreover, HMGB1-dependent autophagy induces chemotherapy resistance in leukemia and multiple myeloma. In this review, we systematically summarize the emerging roles of HMGB1 in carcinogenesis, progression, prognosis, and potential clinical applications in different hematopoietic malignancies. In summary, targeting the regulation of HMGB1 activity in HSCs and the BM microenvironment is highly beneficial in the diagnosis and treatment of various hematopoietic malignancies.
Collapse
Affiliation(s)
- Shunling Yuan
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zhaoping Liu
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zhenru Xu
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Jing Liu
- Hunan Province Key Laboratory of Basic and Applied Hematology, Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China.
| | - Ji Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
17
|
Zhang Y, Zhou K, Wu L, Gu H, Huang Z, Xu J. Downregulation of microRNA‑143 promotes osteogenic differentiation of human adipose‑derived mesenchymal stem cells through the k‑Ras/MEK/ERK signaling pathway. Int J Mol Med 2020; 46:965-976. [PMID: 32582994 PMCID: PMC7388841 DOI: 10.3892/ijmm.2020.4651] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 11/22/2019] [Indexed: 12/29/2022] Open
Abstract
MicroRNAs (miRNAs) are known to have regulatory roles in the osteogenic differentiation of various mesenchymal stem cells (MSCs), although their regulatory role on human adipose‑derived mesenchymal stem cells (hADSCs) remains unclear. The aim of the present study was to investigate the biological function and underlying molecular mechanism of miRNAs in regulating the osteogenic differentiation of hADSCs using microarray assay. hADSCs differentiated into osteoblasts under culture with osteogenic medium, with an increase observed in calcium deposits and alkaline phosphatase activity. The mRNA levels of bone sialoprotein, osteopontin and osteocalcin increased, whereas Runt‑related transcription factor‑2 expression decreased during osteogenic differentiation. In addition, miR‑143 was markedly downregulated during osteogenic differentiation, while miR‑143 overexpression inhibited and miR‑143 knockdown enhanced this process. miR‑143 overexpression also blocked extracellular signal‑regulated kinase 1/2 (ERK1/2) pathway activation, while miR‑143 inhibition enhanced it. The promoting effects of miR‑143 knockdown on the osteogenic differentiation of hADSCs were partly diminished by the mitogen‑activated protein kinase (MEK) inhibitors U0126 and PD98059. Bioinformatics analysis further revealed that miR‑143 targets k‑Ras and directly binds to the 3'‑untranslated region of its mRNA. Inhibition of miR‑143 enhanced the activation of the k‑Ras/MEK/ERK pathway during osteogenic differentiation, whereas miR‑143 overexpression had the opposite effect. Collectively, these results demonstrated that miR‑143 negatively regulates the osteogenic differentiation of hADSCs through the k‑Ras/MEK/ERK pathway, providing further insight into the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Kaifeng Zhou
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Liang Wu
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Huijie Gu
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Zhongyue Huang
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Jun Xu
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| |
Collapse
|
18
|
Wang S, Cai S, Zhang W, Liu X, Li Y, Zhang C, Zeng Y, Xu M, Rong R, Yang T, Shi B, Chandraker A, Yang C, Zhu T. High-mobility group box 1 protein antagonizes the immunosuppressive capacity and therapeutic effect of mesenchymal stem cells in acute kidney injury. J Transl Med 2020; 18:175. [PMID: 32312307 PMCID: PMC7169035 DOI: 10.1186/s12967-020-02334-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/08/2020] [Indexed: 12/13/2022] Open
Abstract
Background Kidney ischemia reperfusion injury (IRI) is a common cause of acute kidney injury and an unavoidable consequence of kidney transplantation and still lacks specific therapeutics. Recently, mesenchymal stem cell (MSC) has been emerging as a promising cell-based therapy for IRI in the context of transplantation. MSC negatively regulates the secretion of pro-inflammatory as well as the activation of immune cells during IRI through its unique immunosuppressive property. Methods We employed mice kidney IRI model and MSC cell line to monitor the IRI related checkpoints. siRNAs were utilized to knock down the potential key factors for mechanistic analysis. Statistical analysis was performed by using one-way ANOVA with Tukey’s post hoc procedure by SPSS. Results The expression of high-mobility group box 1 protein (HMGB1) is increased in the acute phase as well as the recovery stage of IRI. Importantly, the HMGB1 upregulation is correlated with the injury severity. HMGB1 diminishes the MSC induced immunosuppressive capacity in the presence of pro-inflammatory cytokines in vitro. Toll like receptor 4 (TLR4)-mediated inducible nitric oxide synthase (iNOS) inhibition contributes to the negative effect of HMGB1 on MSCs. HMGB1-TLR4 signaling inhibition augments the therapeutic efficacy of MSCs in mice renal IRI model. Conclusions These findings demonstrate that HMGB1 plays a crucial role in shaping the immunoregulatory property of MSCs within the microenvironments, providing novel insights into the crosstalk between MSCs and microenvironment components, suggesting HMGB1 signals as a promising target to improve MSC-based therapy.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China.,Department of Urology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Songjie Cai
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Ave, LRMC 301, Boston, MA, 02115, USA
| | - Weitao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China
| | - Xigao Liu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yan Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Chao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China
| | - Yigang Zeng
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Ming Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China.,Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Tianshu Yang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Anil Chandraker
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Ave, LRMC 301, Boston, MA, 02115, USA.
| | - Cheng Yang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China. .,Fudan Zhangjiang Institute, Shanghai, 201203, China.
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China. .,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
19
|
De Martinis M, Ginaldi L, Sirufo MM, Pioggia G, Calapai G, Gangemi S, Mannucci C. Alarmins in Osteoporosis, RAGE, IL-1, and IL-33 Pathways: A Literature Review. MEDICINA (KAUNAS, LITHUANIA) 2020; 56:medicina56030138. [PMID: 32204562 PMCID: PMC7142770 DOI: 10.3390/medicina56030138] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022]
Abstract
Alarmins are endogenous mediators released by cells following insults or cell death to alert the host’s innate immune system of a situation of danger or harm. Many of these, such as high-mobility group box-1 and 2 (HMGB1, HMGB2) and S100 (calgranulin proteins), act through RAGE (receptor for advanced glycation end products), whereas the IL-1 and IL-33 cytokines bind the IL-1 receptors type I and II, and the cellular receptor ST2, respectively. The alarmin family and their signal pathways share many similarities of cellular and tissue localization, functions, and involvement in various physiological processes and inflammatory diseases including osteoporosis. The aim of the review was to evaluate the role of alarmins in osteoporosis. A bibliographic search of the published scientific literature regarding the role of alarmins in osteoporosis was organized independently by two researchers in the following scientific databases: Pubmed, Scopus, and Web of Science. The keywords used were combined as follows: “alarmins and osteoporosis”, “RAGE and osteoporosis”, “HMGB1 and osteoporosis”, “IL-1 and osteoporosis”, “IL 33 and osteopororsis”, “S100s protein and osteoporosis”. The information was summarized and organized in the present review. We highlight the emerging roles of alarmins in various bone remodeling processes involved in the onset and development of osteoporosis, as well as their potential role as biomarkers of osteoporosis severity and progression. Findings of the research suggest a potential use of alarmins as pharmacological targets in future therapeutic strategies aimed at preventing bone loss and fragility fractures induced by aging and inflammatory diseases.
Collapse
Affiliation(s)
- Massimo De Martinis
- Department of Life, Health, & Environmental Sciences, University of L’Aquila, 6700 L’Aquila, Italy; (M.D.M.); (L.G.); (M.M.S.)
| | - Lia Ginaldi
- Department of Life, Health, & Environmental Sciences, University of L’Aquila, 6700 L’Aquila, Italy; (M.D.M.); (L.G.); (M.M.S.)
| | - Maria Maddalena Sirufo
- Department of Life, Health, & Environmental Sciences, University of L’Aquila, 6700 L’Aquila, Italy; (M.D.M.); (L.G.); (M.M.S.)
| | - Giovanni Pioggia
- National Research Council of Italy (CNR)-Institute for Biomedical Research and Innovation (IRIB), 98164 Messina, Italy;
| | - Gioacchino Calapai
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- School and Division of Allergy and Clinical Immunology, Department of Experimental Medicine, University of Messina, 98125 Messina, Italy;
| | - Carmen Mannucci
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy;
- Correspondence: ; Tel.: +39-090-22-12-697
| |
Collapse
|
20
|
Extracellular IL-37 promotes osteogenic differentiation of human bone marrow mesenchymal stem cells via activation of the PI3K/AKT signaling pathway. Cell Death Dis 2019; 10:753. [PMID: 31582734 PMCID: PMC6776644 DOI: 10.1038/s41419-019-1904-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 07/24/2019] [Accepted: 08/11/2019] [Indexed: 12/11/2022]
Abstract
Interleukin (IL)-37, a pivotal anti-inflammatory cytokine and a fundamental inhibitor of innate immunity, has recently been shown to be abnormally expressed in several autoimmune-related orthopedic diseases, including rheumatoid arthritis, ankylosing spondylitis, and osteoporosis. However, the role of IL-37 during osteogenic differentiation of mesenchymal stem cells (MSCs) remains largely unknown. In this study, extracellular IL-37 significantly increased osteoblast-specific gene expression, the number of mineral deposits, and alkaline phosphatase activity of MSCs. Moreover, a signaling pathway was activated in the presence of IL-37. The enhanced osteogenic differentiation of MSCs due to supplementation of IL-37 was partially rescued by the presence of a PI3K/AKT signaling inhibitor. Using a rat calvarial bone defect model, IL-37 significantly improved bone healing. Collectively, these findings indicate that extracellular IL-37 enhanced osteogenesis of MSCs, at least in part by activation of the PI3K/AKT signaling pathway.
Collapse
|
21
|
Xu Y, An JJ, Tabys D, Xie YD, Zhao TY, Ren HW, Liu N. Effect of Lactoferrin on the Expression Profiles of Long Non-coding RNA during Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells. Int J Mol Sci 2019; 20:ijms20194834. [PMID: 31569432 PMCID: PMC6801644 DOI: 10.3390/ijms20194834] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/21/2019] [Accepted: 09/25/2019] [Indexed: 12/13/2022] Open
Abstract
Lactoferrin (LF) has demonstrated stimulation of osteogenic differentiation of mesenchymal stem cells (MSCs). Long non-coding RNAs (lncRNAs) participate in regulating the osteogenic differentiation processes. However, the impact of LF on lncRNA expression in MSC osteogenic differentiation is poorly understood. Our aim was to investigate the effects of LF on lncRNAs expression profiles, during osteogenic differentiation of rat bone marrow mesenchymal stem cells (rBMSCs), by RNA sequencing. A total number of 1331 putative lncRNAs were identified in rBMSCs during osteogenic differentiation in the study. LF influenced the expression of 120 lncRNAs (differentially expressed lncRNAs [DELs], Fold change > 1.5 or < −1.5; p < 0.05) in rBMSCs on day 14 of osteogenic differentiation, consisted of 60 upregulated and 60 down-regulated. Furthermore, the potential functions of DELs were of prediction by searching their target cis- and trans-regulated protein-coding genes. The bioinformatic analysis of DELs target gene revealed that LF led to the disfunction of transforming growth factor beta stimulus (TGF-β) and positive regulation of I-κappa B kinase/NF-κappa B signaling pathway, which may relate to osteogenic differentiation of rBMSCs. Our work is the first profiling of lncRNA in osteogenic differentiation of rBMSCs induced by LF, and provides valuable insights into the potential mechanisms for LF promoting osteogenic activity.
Collapse
Affiliation(s)
- Yan Xu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
- College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| | - Jing-Jing An
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
- College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| | - Dina Tabys
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
- College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| | - Yin-Dan Xie
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
- College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| | - Tian-Yu Zhao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
- College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| | - Hao-Wei Ren
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
- College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| | - Ning Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
- College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
22
|
Chen MQ, Luan JJ. HMGB1 promotes bone fracture healing through activation of ERK signaling pathway in a rat tibial fracture model. Kaohsiung J Med Sci 2019; 35:550-558. [PMID: 31162822 DOI: 10.1002/kjm2.12095] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 05/12/2019] [Indexed: 01/04/2023] Open
Abstract
This work was to investigate potential roles of HMGB1-mediated ERK pathway in the healing process of bone fracture. Rat tibial fracture models were established and divided into control (rats with normal saline), HMGB1 (rats with HMGB1), and HMGB1+ PD98059 groups (rats with HMGB1 and 1 mg/kg of ERK1/2 inhibitor PD98059) with 30 rats per each. The healing of rats' fracture was observed by X-ray films, the morphological changes of bone fractures by HE staining, the callus formation by micro-CT and biomechanical test, and the expression of osteogenesis-related genes, HMGB1 and ERK-related proteins by qRT-PCR and Western blot. Rats in the HMGB1 group was increased in X-ray scores, peak torque, torsional stiffness, and the bone volume fraction (bone volume/total volume, BV/TV); meanwhile, those rats presented elevations in osteogenesis-related genes and HMGB1 expressions, as well as p-ERK/ERK ratio. However, rats in the HMGB1+ PD98059 group was significantly reduced in X-ray score, peak torque, torsional stiffness, and BV/TV, as well as the expression of osteogenesis-related genes and the ratio of p-ERK/ERK, as compared to those from HMGB1 group. HMGB1 could promote the expressions of osteogenesis-related genes and accelerate the healing process of fracture via activation of the ERK signaling pathway.
Collapse
Affiliation(s)
- Ming-Qi Chen
- Department of Traumatic Orthopedics, YanTaiShan Hospital, YanTai City, Shandong, China
| | - Jing-Jie Luan
- Department of Traumatic Orthopedics, YanTaiShan Hospital, YanTai City, Shandong, China
| |
Collapse
|
23
|
Davis HM, Valdez S, Gomez L, Malicky P, White FA, Subler MA, Windle JJ, Bidwell JP, Bruzzaniti A, Plotkin LI. High mobility group box 1 protein regulates osteoclastogenesis through direct actions on osteocytes and osteoclasts in vitro. J Cell Biochem 2019; 120:16741-16749. [PMID: 31106449 DOI: 10.1002/jcb.28932] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/24/2019] [Indexed: 12/30/2022]
Abstract
Old age and Cx43 deletion in osteocytes are associated with increased osteocyte apoptosis and osteoclastogenesis. We previously demonstrated that apoptotic osteocytes release elevated concentrations of the proinflammatory cytokine, high mobility group box 1 protein (HMGB1) and apoptotic osteocyte conditioned media (CM) promotes osteoclast differentiation. Further, prevention of osteocyte apoptosis blocks osteoclast differentiation and attenuates the extracellular release of HMGB1 and RANKL. Moreover, sequestration of HMGB1, in turn, reduces RANKL production/release by MLO-Y4 osteocytic cells silenced for Cx43 (Cx43def ), highlighting the possibility that HMGB1 promotes apoptotic osteocyte-induced osteoclastogenesis. However, the role of HMGB1 signaling in osteocytes has not been well studied. Further, the mechanisms underlying its release and the receptor(s) responsible for its actions is not clear. We now report that a neutralizing HMGB1 antibody reduces osteoclast formation in RANKL/M-CSF treated bone marrow cells. In bone marrow macrophages (BMMs), toll-like receptor 4 (TLR4) inhibition with LPS-RS, but not receptor for advanced glycation end products (RAGE) inhibition with Azeliragon attenuated osteoclast differentiation. Further, inhibition of RAGE but not of TLR4 in osteoclast precursors reduced osteoclast number, suggesting that HGMB1 produced by osteoclasts directly affects differentiation by activating TLR4 in BMMs and RAGE in preosteoclasts. Our findings also suggest that increased osteoclastogenesis induced by apoptotic osteocytes CM is not mediated through HMGB1/RAGE activation and that direct HMGB1 actions in osteocytes stimulate pro-osteoclastogenic signal release from Cx43def osteocytes. Based on these findings, we propose that HMGB1 exerts dual effects on osteoclasts, directly by inducing differentiation through TLR4 and RAGE activation and indirectly by increasing pro-osteoclastogenic cytokine secretion from osteocytes.
Collapse
Affiliation(s)
- Hannah M Davis
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana.,Indiana Center for Musculoskeletal Health, Indianapolis, Indiana
| | - Sinai Valdez
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Leland Gomez
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Peter Malicky
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, Indiana
| | - Fletcher A White
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, Indiana.,Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana.,Roudebush Veterans Administration Medical Center, Indianapolis, Indiana
| | - Mark A Subler
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia
| | - Jolene J Windle
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia
| | - Joseph P Bidwell
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana.,Indiana Center for Musculoskeletal Health, Indianapolis, Indiana
| | - Angela Bruzzaniti
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana.,Indiana Center for Musculoskeletal Health, Indianapolis, Indiana.,Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Lilian I Plotkin
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana.,Indiana Center for Musculoskeletal Health, Indianapolis, Indiana.,Roudebush Veterans Administration Medical Center, Indianapolis, Indiana
| |
Collapse
|
24
|
Biguetti CC, Cavalla F, Silveira EV, Tabanez AP, Francisconi CF, Taga R, Campanelli AP, Trombone APF, Rodrigues DC, Garlet GP. HGMB1 and RAGE as Essential Components of Ti Osseointegration Process in Mice. Front Immunol 2019; 10:709. [PMID: 31024546 PMCID: PMC6461067 DOI: 10.3389/fimmu.2019.00709] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/15/2019] [Indexed: 12/31/2022] Open
Abstract
The release of the prototypic DAMP High Mobility Group Box 1 (HMGB1) into extracellular environment and its binding to the Receptor for Advanced Glycation End Products (RAGE) has been described to trigger sterile inflammation and regulate healing outcome. However, their role on host response to Ti-based biomaterials and in the subsequent osseointegration remains unexplored. In this study, HMGB1 and RAGE inhibition in the Ti-mediated osseointegration were investigated in C57Bl/6 mice. C57Bl/6 mice received a Ti-device implantation (Ti-screw in the edentulous alveolar crest and a Ti-disc in the subcutaneous tissue) and were evaluated by microscopic (microCT [bone] and histology [bone and subcutaneous]) and molecular methods (ELISA, PCR array) during 3, 7, 14, and 21 days. Mice were divided into 4 groups: Control (no treatment); GZA (IP injection of Glycyrrhizic Acid for HMGB1 inhibition, 4 mg/Kg/day); RAP (IP injection of RAGE Antagonistic Peptide, 4 mg/Kg/day), and vehicle controls (1.5% DMSO solution for GZA and 0.9% saline solution for RAP); treatments were given at all experimental time points, starting 1 day before surgeries. HMGB1 was detected in the Ti-implantation sites, adsorbed to the screws/discs. In Control and vehicle groups, osseointegration was characterized by a slight inflammatory response at early time points, followed by a gradual bone apposition and matrix maturation at late time points. The inhibition of HMGB1 or RAGE impaired the osseointegration, affecting the dynamics of mineralized and organic bone matrix, and resulting in a foreign body reaction, with persistence of macrophages, necrotic bone, and foreign body giant cells until later time points. While Control samples were characterized by a balance between M1 and M2-type response in bone and subcutaneous sites of implantation, and also MSC markers, the inhibition of HMGB1 or RAGE caused a higher expression M1 markers and pro-inflammatory cytokines, as well chemokines and receptors for macrophage migration until later time points. In conclusion, HMGB1 and RAGE have a marked role in the osseointegration, evidenced by their influence on host inflammatory immune response, which includes macrophages migration and M1/M2 response, MSC markers expression, which collectively modulate bone matrix deposition and osseointegration outcome.
Collapse
Affiliation(s)
- Claudia Cristina Biguetti
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Franco Cavalla
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, São Paulo, Brazil.,Department of Conservative Dentistry, School of Dentistry, University of Chile, Santiago, Chile
| | - Elcia Varize Silveira
- Department of Biological and Allied Health Sciences, Universidade Sagrado Coração, Bauru, Brazil
| | - André Petenuci Tabanez
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, São Paulo, Brazil
| | | | - Rumio Taga
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Ana Paula Campanelli
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, São Paulo, Brazil
| | | | - Danieli C Rodrigues
- Department of Bioengineering, University of Texas at Dallas, Dallas, TX, United States
| | - Gustavo Pompermaier Garlet
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
25
|
Tao X, Sun M, Chen M, Ying R, Su W, Zhang J, Xie X, Wei W, Meng X. HMGB1-modified mesenchymal stem cells attenuate radiation-induced vascular injury possibly via their high motility and facilitation of endothelial differentiation. Stem Cell Res Ther 2019; 10:92. [PMID: 30867070 PMCID: PMC6416980 DOI: 10.1186/s13287-019-1197-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 12/28/2022] Open
Abstract
Background Vascular injury is one of the most common detrimental effects of cancer radiotherapy on healthy tissues. Since the efficacy of current preventive and therapeutic strategies remains limited, the exploration of new approaches to treat radiation-induced vascular injury (RIV) is on high demands. The use of mesenchymal stem cells (MSCs) to treat RIV holds great promise thanks to their well-documented function of mediating tissue regeneration after injury. Recently, we genetically modified MSCs with high mobility group box 1 (HMGB1) and demonstrated the high efficacy of these cells in treating graft atherosclerosis. The current study was to investigate the protective effect of HMGB1-modified MSCs (MSC-H) on RIV by using a rat model. Methods Female F344 rats received an intravenous injection of male F344 MSC-H cells or vehicle control at four doses of 2 × 106 cells with a 15-day interval starting from 30 days after irradiation to the abdominal aorta. The aortas were procured for histological and biomedical analysis at 90 days after irradiation. Cell migration to irradiated aortas was traced by green fluorescent protein and sex determination region on the Y chromosome. In vitro cell migration and endothelial differentiation of MSC-H cells were analyzed by stromal-derived factor 1-induced transwell assay and RNA microarray, respectively. The contribution of extracellular HMGB1 to the bioactivity of MSC-H cells was investigated by inhibition experiments with HMGB1 antibody. Result MSC-H cell infusion alleviated neointimal formation, vascular inflammation, and fibrosis in irradiated aortas, which was associated with local migration and endothelial differentiation of MSC-H cells. The MSC-H cells showed high motility and potential of endothelial differentiation in vitro. Microarray analysis suggested multiple pathways like MAPK and p53 signaling were activated during endothelial differentiation. MSC-H cells highly expressed CXC chemokine receptor 4 and migrated progressively after stromal-derived factor 1 stimulation, which was blocked by the antagonist of CXC chemokine receptor 4. Finally, the migration and endothelial differentiation of MSC-H cells were inhibited by HMGB1 antibody. Conclusion MSC-H cell infusion significantly attenuated RIV, which was associated with their high motility and endothelial differentiation potential. Multiple pathways that possibly contributed to the efficacy of MSC-H cells were suggested and deserved further investigation. Electronic supplementary material The online version of this article (10.1186/s13287-019-1197-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xuan Tao
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mingyang Sun
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Chen
- Department of Gastroenterology, Nanjing University Medical School, Nanjing Drum Tower Hospital, Nanjing, China
| | - Rongchao Ying
- Department of Gastroenterological Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjie Su
- Department of Gastroenterological Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Zhang
- Department of Gastroenterological Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaodong Xie
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Wei
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China. .,Department of Gastroenterological Surgery, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, China.
| | - Xiaohu Meng
- Division of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
26
|
Li C, Wang D, Guan X, Liu S, Su P, Li Q, Pang Y. HMGB1 from Lampetra japonica promotes inflammatory activation in supraneural body cells. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 92:50-59. [PMID: 30423344 DOI: 10.1016/j.dci.2018.11.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 11/08/2018] [Accepted: 11/08/2018] [Indexed: 06/09/2023]
Abstract
High mobility group box protein 1 (HMGB1) acts as a potent proinflammatory cytokine that involves in the pathogenesis of diverse inflammatory and infectious disorders. In previous study, we identified a homolog of HMGB1 in the Lampetra japonica(L-HMGB1), and further revealed that L-HMGB1 was able to induce the production of tumor necrosis factor-α (TNF-α) in activated human acute monocytic leukemia cells. However, the role of L-HMGB1 played in lamprey was unknown. Here, we found that L-HMGB1 was located in the cytoplasm of lamprey leukocytes and supraneural body (SB) cells. Importantly, we demonstrated that L-HMGB1 participated in activation of various key molecules in inflammation signaling pathway. LPS also promoted the release of L-HMGB1 from SB cells similar to Hu-HMGB1, and then extracellular L-HMGB1 in turn induced the release of cytokines. This study revealed that the synergistic action of LPS and L-HMGB1 played a crucial role in inflammation in lamprey. Our results suggested that lampreys used L-HMGB1 to activate their innate immunity for the purpose of pathogen defense.
Collapse
Affiliation(s)
- Changzhi Li
- College of Life Science, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Dong Wang
- College of Life Science, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Xin Guan
- College of Life Science, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Shuang Liu
- College of Life Science, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Peng Su
- College of Life Science, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Qingwei Li
- College of Life Science, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| | - Yue Pang
- College of Life Science, Liaoning Normal University, Dalian, 116081, China; Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW The receptor for advanced glycation end products (RAGE) and several of its ligands have been implicated in the onset and progression of pathologies associated with aging, chronic inflammation, and cellular stress. In particular, the role of RAGE and its ligands in bone tissue during both physiological and pathological conditions has been investigated. However, the extent to which RAGE signaling regulates bone homeostasis and disease onset remains unclear. Further, RAGE effects in the different bone cells and whether these effects are cell-type specific is unknown. The objective of the current review is to describe the literature over RAGE signaling in skeletal biology as well as discuss the clinical potential of RAGE as a diagnostic and/or therapeutic target in bone disease. RECENT FINDINGS The role of RAGE and its ligands during skeletal homeostasis, tissue repair, and disease onset/progression is beginning to be uncovered. For example, detrimental effects of the RAGE ligands, advanced glycation end products (AGEs), have been identified for osteoblast viability/activity, while others have observed that low level AGE exposure stimulates osteoblast autophagy, which subsequently promotes viability and function. Similar findings have been reported with HMGB1, another RAGE ligand, in which high levels of the ligand are associated with osteoblast/osteocyte apoptosis, whereas low level/short-term administration stimulates osteoblast differentiation/bone formation and promotes fracture healing. Additionally, elevated levels of several RAGE ligands (AGEs, HMGB1, S100 proteins) induce osteoblast/osteocyte apoptosis and stimulate cytokine production, which is associated with increased osteoclast differentiation/activity. Conversely, direct RAGE-ligand exposure in osteoclasts may have inhibitory effects. These observations support a conclusion that elevated bone resorption observed in conditions of high circulating ligands and RAGE expression are due to actions on osteoblasts/osteocytes rather than direct actions on osteoclasts, although additional work is required to substantiate the observations. Recent studies have demonstrated that RAGE and its ligands play an important physiological role in the regulation of skeletal development, homeostasis, and repair/regeneration. Conversely, elevated levels of RAGE and its ligands are clearly related with various diseases associated with increased bone loss and fragility. However, despite the recent advancements in the field, many questions regarding RAGE and its ligands in skeletal biology remain unanswered.
Collapse
Affiliation(s)
- Lilian I Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr., MS 5023, Indianapolis, IN, 46202, USA.
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.
- Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA.
| | - Alyson L Essex
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr., MS 5023, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA
| | - Hannah M Davis
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr., MS 5023, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA
| |
Collapse
|
28
|
Zhang Z, Li Z, Zhang C, Liu J, Bai Y, Li S, Zhang C. Biomimetic intrafibrillar mineralized collagen promotes bone regeneration via activation of the Wnt signaling pathway. Int J Nanomedicine 2018; 13:7503-7516. [PMID: 30538446 PMCID: PMC6257138 DOI: 10.2147/ijn.s172164] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose The purpose of this study was to assess the effects of biomimetic intrafibrillar mineralized collagen (IMC) bone scaffold materials on bone regeneration and the underlying biological mechanisms. Materials and methods A critical-sized bone defect in the rat femur was created; then IMC, extrafibrillar mineralized collagen, and nano-hydroxyapatite bone scaffold materials were grafted into the defect. Ten weeks after implantation, micro-computed tomography and histology were applied to evaluate the bone regeneration. Furthermore, microarray technology was applied for transcriptional profile analysis at two postoperative time points (7 and 14 days). Subsequently, the critical genes involved in bone regeneration identified by transcriptional analysis were verified both in vivo through immunohistochemical analysis and in vitro by quantitative real-time transcription polymerase chain reaction evaluation. Results Significantly increased new bone formation was found in the IMC group based on micro-computed tomography and histological evaluation (P<0.05). Transcriptional analysis revealed that the early process of IMC-guided bone regeneration involves the overexpression of genes mainly associated with inflammation, immune response, skeletal development, angiogenesis, neurogenesis, and the Wnt signaling pathway. The roles of the Wnt signaling pathway-related factors Wnt5a, β-catenin, and Axin2 were further confirmed both in vivo and in vitro. Conclusion The IMC bone scaffold materials significantly enhanced bone regeneration via activation of the Wnt signaling pathway.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research, Institute of Stomatology and National Clinical Research Center of Stomatology, Shanghai, China,
| | - Zheyi Li
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China, .,Institute for Clinical Research and Application of Sunny Dental, Beijing, China
| | - Chengyao Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research, Institute of Stomatology and National Clinical Research Center of Stomatology, Shanghai, China,
| | - Jiannan Liu
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research, Institute of Stomatology and National Clinical Research Center of Stomatology, Shanghai, China,
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China,
| | - Song Li
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China,
| | - Chenping Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research, Institute of Stomatology and National Clinical Research Center of Stomatology, Shanghai, China,
| |
Collapse
|
29
|
Rayavara K, Kurosky A, Stafford SJ, Garg NJ, Brasier AR, Garofalo RP, Hosakote YM. Proinflammatory Effects of Respiratory Syncytial Virus-Induced Epithelial HMGB1 on Human Innate Immune Cell Activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2753-2766. [PMID: 30275049 PMCID: PMC6200588 DOI: 10.4049/jimmunol.1800558] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/29/2018] [Indexed: 01/21/2023]
Abstract
High mobility group box 1 (HMGB1) is a multifunctional nuclear protein that translocates to the cytoplasm and is subsequently released to the extracellular space during infection and injury. Once released, it acts as a damage-associated molecular pattern and regulates immune and inflammatory responses. Respiratory syncytial virus (RSV) is a major cause of acute lower respiratory tract infections in infants and elderly, for which no effective treatment or vaccine is currently available. This study investigated the effects of HMGB1 on cytokine secretion, as well as the involvement of NF-κB and TLR4 pathways in RSV-induced HMGB1 release in human airway epithelial cells (AECs) and its proinflammatory effects on several human primary immune cells. Purified HMGB1 was incubated with AECs (A549 and small alveolar epithelial cells) and various immune cells and measured the release of proinflammatory mediators and the activation of NF-κB and P38 MAPK. HMGB1 treatment significantly increased the phosphorylation of NF-κB and P38 MAPK but did not induce the release of cytokines/chemokines from AECs. However, addition of HMGB1 to immune cells did significantly induce the release of cytokines/chemokines and activated the NF-κB and P38 MAPK pathways. We found that activation of NF-κB accounted for RSV-induced HMGB1 secretion in AECs in a TLR4-dependent manner. These results indicated that HMGB1 secreted from AECs can facilitate the secretion of proinflammatory mediators from immune cells in a paracrine mechanism, thus promoting the inflammatory response that contributes to RSV pathogenesis. Therefore, blocking the proinflammatory function of HMGB1 may be an effective approach for developing novel therapeutics.
Collapse
Affiliation(s)
- Kempaiah Rayavara
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77555
| | - Alexander Kurosky
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77555
| | - Susan J Stafford
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77555
| | - Nisha J Garg
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77555
| | - Allan R Brasier
- Division of Endocrinology, Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX 77555
- Institute for Translational Sciences, The University of Texas Medical Branch, Galveston, TX 77555
| | - Roberto P Garofalo
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555; and
- Sealy Center for Vaccine Development, The University of Texas Medical Branch, Galveston, TX 77555
| | - Yashoda M Hosakote
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77555;
- Institute for Translational Sciences, The University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
30
|
Lin F, Zhu Y, Hu G. Naringin promotes cellular chemokine synthesis and potentiates mesenchymal stromal cell migration via the Ras signaling pathway. Exp Ther Med 2018; 16:3504-3510. [PMID: 30233702 PMCID: PMC6143896 DOI: 10.3892/etm.2018.6634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 07/13/2018] [Indexed: 12/17/2022] Open
Abstract
Directional migration of mesenchymal stem cells (MSCs) is known to serve roles in bone fracture healing. Naringin is a traditional medicine used in China to treat bone injury and has been confirmed to act as a chemoattractant to MSCs. In the present study, the secretion of chemokines and stimulation of relevant signaling pathways by naringin were detected to determine the molecular mechanism of naringin-induced MSC migration. In these experiments, Quantibody® arrays were used to detect chemokines secreted by MSCs with or without the addition of naringin. The results revealed differential naringin-induced chemokine secretion of C-X-C motif chemokine (CXCL)5, CXCL6 and C-C motif chemokine 20. Furthermore, the Ras signaling pathway was markedly activated in the naringin-treated groups, suggesting that naringin may enhance the migrational ability of MSCs via Ras activation. Furthermore, naringin was able to promote the secretion of various chemokines derived from MSCs, which would, in turn, increase the mobility of MSCs. The aim of the present study was to provide novel candidate agents for clinical orthopedics and theoretical basis for the future improvement of adjunctive medication for bone fracture healing.
Collapse
Affiliation(s)
- Feng Lin
- Department of Orthopedics, Xiaoshan First People's Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Yuan Zhu
- Department of Orthopedics, Xiaoshan First People's Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Gangfeng Hu
- Department of Orthopedics, Xiaoshan First People's Hospital, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
31
|
Gefitinib Inhibits Bleomycin-Induced Pulmonary Fibrosis via Alleviating the Oxidative Damage in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8249693. [PMID: 29849916 PMCID: PMC5924979 DOI: 10.1155/2018/8249693] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 10/24/2017] [Accepted: 11/26/2017] [Indexed: 01/09/2023]
Abstract
Pulmonary fibrosis (PF) is a life-threatening interstitial lung disease. In this study, we tried to reveal the model of action between high-mobility group box 1 (HMGB1) and α-smooth muscle actin (α-SMA) and the protective role of gefitinib in pulmonary fibrosis induced by the administration of bleomycin aerosol in mice. For the mechanism study, lung tissues were harvested two weeks after modeling to detect the coexpression of HMGB1 and α-SMA by immunohistochemistry and immunofluorescence staining. Protein-DNA interactions were analyzed using a pulldown assay to study the relationship between HMGB1 and α-SMA. For the gefitinib treatment study, the mice were divided into three groups: phosphate-buffered saline (PBS) control group, PBS-treated PF group, and gefitinib-treated PF group. Gavage of gefitinib or PBS (20 mg/kg/day) was performed after bleomycin treatment for two weeks until the mice were sacrificed. Lung and blood samples were collected to assess the histological changes, oxidative stress, and expression of NOXs, HMGB1, EGFR, MAPKs, AP-1, and NF-κB to determine the curative effect and related molecular mechanisms. The results revealed the high coexpression of α-SMA and HMGB1 in some interstitial cells in the fibrotic lung. The DNA-protein pulldown analysis proved that HMGB34367 acted as a novel transcriptional factor for the α-SMA promoter and participated in the eventual development of pulmonary fibrosis. Second, gefitinib could significantly decrease lung fibrotic changes and the level of MDA and recover the T-AOC level. Meanwhile, gefitinib could also reduce the NOX1/2/4, HMGB1, p-EGFR, p-ERK, p-JNK, p-P38, p-NF-κB, p-c-Jun, and p-c-Fos expression levels in fibrotic lungs. The present study suggested that gefitinib could alleviate lung fibrosis through the HMGB1/NOXs-ROS/EGFR-MAPKs-AP-1/NF-κB signal in bleomycin-induced pulmonary fibrosis.
Collapse
|
32
|
Chen E, Liu G, Zhou X, Zhang W, Wang C, Hu D, Xue D, Pan Z. Concentration-dependent, dual roles of IL-10 in the osteogenesis of human BMSCs via P38/MAPK and NF-κB signaling pathways. FASEB J 2018; 32:4917-4929. [PMID: 29630408 DOI: 10.1096/fj.201701256rrr] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Microenvironmental conditions can influence the differentiation and functional roles of mesenchymal stem cells (MSCs). Recent studies have suggested that an inflammatory microenvironment can significantly affect the osteogenic differentiation of MSCs. Here, we show, for the first time, that IL-10 has concentration-dependent, dual roles in the osteogenesis of human bone marrow mesenchymal stem cells (hBMSCs). Low physiologic concentrations of IL-10 (0.01-1.0 ng/ml) activate the p38/MAPK signaling pathway to promote the osteogenesis of hBMSCs, but higher pathologic doses of IL-10 (10-100 ng/ml) inhibit p38/MAPK signaling by activating NF-κB, inhibiting osteogenesis. These results demonstrate that p38/MAPK and NF-κB signaling mediates the double-edged sword effect of IL-10 on hBMSCs. The osteogenic impairment was reversed at higher doses of IL-10 when cells were supplemented with the NF-κB inhibitor BAY11-7082. These data provide important insights into the regulatory effects of IL-10 on the biologic behavior of hBMSCs.-Chen, E., Liu, G., Zhou, X., Zhang, W., Wang, C., Hu, D., Xue, D., Pan, Z. Concentration-dependent, dual roles of IL-10 in the osteogenesis of human BMSCs via P38/MAPK and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Erman Chen
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; and.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Guanyi Liu
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; and.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Xiaopeng Zhou
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; and.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Wei Zhang
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; and.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Cong Wang
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; and
| | - Dongcai Hu
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; and
| | - Deting Xue
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; and.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Zhijun Pan
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; and.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| |
Collapse
|
33
|
Meng X, Chen M, Su W, Tao X, Sun M, Zou X, Ying R, Wei W, Wang B. The differentiation of mesenchymal stem cells to vascular cells regulated by the HMGB1/RAGE axis: its application in cell therapy for transplant arteriosclerosis. Stem Cell Res Ther 2018; 9:85. [PMID: 29615103 PMCID: PMC5883535 DOI: 10.1186/s13287-018-0827-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/18/2018] [Accepted: 03/06/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC) transplantation shows promise for treating transplant arteriosclerosis, at least partly via promoting endothelial regeneration. However, the efficacy and safety are still under investigation especially regarding recent findings that neointimal smooth muscle cells are derived from MSC-like cells. The high mobility group box 1 (HMGB1)/receptor for advanced glycation end-product (RAGE) axis is involved in regulating proliferation, migration, and differentiation of MSCs, and therefore it can be presumably applied to improve the outcome of cell therapy. The aim of the current study was to investigate this hypothesis. METHODS Rat MSCs were treated with HMGB1 or modified with HMGB1 vectors to activate the HMGB1/RAGE axis. RAGE was targeted and inhibited by specific short hairpin RNA vectors. We assessed the capacity for cell proliferation, migration, and differentiation after vector transfection in vitro and in a rat model of transplant arteriosclerosis. The expression of CD31 and α-smooth muscle actin (αSMA) was determined to evaluate the differentiation of MSCs to endothelial cells and smooth muscle cells. RESULTS Exogenous HMGB1 treatment and transfection with HMGB1 vectors promoted MSC migration and vascular endothelial growth factor (VEGF)-induced differentiation to CD31+ cells while inhibiting their proliferation and platelet-derived growth factor (PDGF)-induced differentiation to αSMA+ cells. Such an effect was blocked by RAGE knockdown. HMGB1-modified cells preferably migrated to graft neointima and differentiated to CD31+ cells along with significant relief of transplant arteriosclerosis and inhibition of HMGB1 and RAGE expression in graft vessels. RAGE knockdown inhibited cell migration to graft vessels. CONCLUSIONS HMGB1 stimulated MSCs to migrate and differentiate to endothelial cells via RAGE signaling, which we translated to successful application in cell therapy for transplant arteriosclerosis.
Collapse
Affiliation(s)
- Xiaohu Meng
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan, Nanjing, 210011, China
| | - Min Chen
- Department of Gastroenterology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Wenjie Su
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital Affiliated to Nanjing Medical University, Hangzhou, China
| | - Xuan Tao
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan, Nanjing, 210011, China
| | - Mingyang Sun
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan, Nanjing, 210011, China
| | - Xiaoping Zou
- Department of Gastroenterology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Rongchao Ying
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital Affiliated to Nanjing Medical University, Hangzhou, China
| | - Wei Wei
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan, Nanjing, 210011, China.
| | - Baolin Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan, Nanjing, 210011, China.
| |
Collapse
|
34
|
Aoyagi H, Yamashiro K, Hirata‐Yoshihara C, Ideguchi H, Yamasaki M, Kawamura M, Yamamoto T, Kochi S, Wake H, Nishibori M, Takashiba S. HMGB1‐induced inflammatory response promotes bone healing in murine tooth extraction socket. J Cell Biochem 2018; 119:5481-5490. [DOI: 10.1002/jcb.26710] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/23/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Hiroaki Aoyagi
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Keisuke Yamashiro
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Chiaki Hirata‐Yoshihara
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Hidetaka Ideguchi
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Mutsuyo Yamasaki
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Mari Kawamura
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Tadashi Yamamoto
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Shinsuke Kochi
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Hidenori Wake
- Department of PharmacologyOkayama University Graduate School of MedicineDentistry and Pharmacological SciencesOkayamaJapan
| | - Masahiro Nishibori
- Department of PharmacologyOkayama University Graduate School of MedicineDentistry and Pharmacological SciencesOkayamaJapan
| | - Shogo Takashiba
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| |
Collapse
|
35
|
Global phenotypic characterisation of human platelet lysate expanded MSCs by high-throughput flow cytometry. Sci Rep 2018; 8:3907. [PMID: 29500387 PMCID: PMC5834600 DOI: 10.1038/s41598-018-22326-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 02/21/2018] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are a promising cell source to develop cell therapy for many diseases. Human platelet lysate (PLT) is increasingly used as an alternative to foetal calf serum (FCS) for clinical-scale MSC production. To date, the global surface protein expression of PLT-expended MSCs (MSC-PLT) is not known. To investigate this, paired MSC-PLT and MSC-FCS were analysed in parallel using high-throughput flow cytometry for the expression of 356 cell surface proteins. MSC-PLT showed differential surface protein expression compared to their MSC-FCS counterpart. Higher percentage of positive cells was observed in MSC-PLT for 48 surface proteins, of which 13 were significantly enriched on MSC-PLT. This finding was validated using multiparameter flow cytometry and further confirmed by quantitative staining intensity analysis. The enriched surface proteins are relevant to increased proliferation and migration capacity, as well as enhanced chondrogenic and osteogenic differentiation properties. In silico network analysis revealed that these enriched surface proteins are involved in three distinct networks that are associated with inflammatory responses, carbohydrate metabolism and cellular motility. This is the first study reporting differential cell surface protein expression between MSC-PLT and MSC-FSC. Further studies are required to uncover the impact of those enriched proteins on biological functions of MSC-PLT.
Collapse
|
36
|
miR-195 inhibited abnormal activation of osteoblast differentiation in MC3T3-E1 cells via targeting RAF-1. Exp Cell Res 2018; 362:293-301. [DOI: 10.1016/j.yexcr.2017.11.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 01/11/2023]
|
37
|
Fahmy-Garcia S, van Driel M, Witte-Buoma J, Walles H, van Leeuwen JPTM, van Osch GJVM, Farrell E. NELL-1, HMGB1, and CCN2 Enhance Migration and Vasculogenesis, But Not Osteogenic Differentiation Compared to BMP2. Tissue Eng Part A 2017; 24:207-218. [PMID: 28463604 DOI: 10.1089/ten.tea.2016.0537] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Currently, autografts still represent the gold standard treatment for the repair of large bone defects. However, these are associated with donor-site morbidity and increased pain, cost, and recovery time. The ideal therapy would use biomaterials combined with bone growth factors to induce and instruct bone defect repair without the need to harvest patient tissue. In this line, bone morphogenetic proteins (BMPs) have been the most extensively used agents for clinical bone repair, but at supraphysiological doses that are not without risk. Because of the need to eliminate the risks of BMP2 use in vivo, we assessed the ability of three putative osteogenic factors, nel-like molecule type 1 (NELL-1), high mobility group box 1 (HMGB1), and CCN2, to enhance the essential processes for bone defect repair in vitro and compared them to BMP2. Although it has been reported that NELL-1, HMGB1, and CCN2 play a role in bone formation, less is known about the contribution of these proteins to the different events involved, such as cell migration, osteogenesis, and vasculogenesis. In this study, we investigated the effects of different doses of NELL-1, HMGB, CCN2, and BMP2 on these three processes as a model for the recruitment and differentiation of resident cells in the in vivo bone defect repair situation, using cells of human origin. Our data demonstrated that NELL-1, HMGB1, and CCN2 significantly induced mesenchymal stem cell migration (from 1.58-fold increase compared to control), but BMP2 did not. Interestingly, only BMP2 increased osteogenesis in marrow stromal cells, whereas it inhibited osteogenesis in preosteoblasts. Moreover, the four proteins studied promoted significantly endothelial cell migration, reaching a maximum of 2.4-fold increase compared to control, and induced formation of tube-like structures. NELL-1, HMGB1, and CCN2 had these effects at relatively low doses compared to BMP2. This work indicates that NELL-1, HMGB1, and CCN2 might enhance bone defect healing via the recruitment of endogenous cells and induction of vascularization and act via different processes than BMP2.
Collapse
Affiliation(s)
| | | | - Janneke Witte-Buoma
- 3 Department of Oral and Maxillofacial Surgery, Erasmus MC , Rotterdam, The Netherlands
| | - Heike Walles
- 4 Department Tissue Engineering and Regenerative Medicine, University Hospital Würzburg , Würzburg, Germany
| | | | - Gerjo J V M van Osch
- 1 Department of Orthopaedics, Erasmus MC , Rotterdam, The Netherlands .,5 Otorhinolaryngology Department, Erasmus MC, Rotterdam, The Netherlands
| | - Eric Farrell
- 3 Department of Oral and Maxillofacial Surgery, Erasmus MC , Rotterdam, The Netherlands
| |
Collapse
|
38
|
Chu Y, Wang Y, Zheng Z, Lin Y, He R, Liu J, Yang X. Proinflammatory Effect of High Glucose Concentrations on HMrSV5 Cells via the Autocrine Effect of HMGB1. Front Physiol 2017; 8:762. [PMID: 29033853 PMCID: PMC5627536 DOI: 10.3389/fphys.2017.00762] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/19/2017] [Indexed: 12/27/2022] Open
Abstract
Background: Peritoneal fibrosis, in which inflammation and apoptosis play crucial pathogenic roles, is a severe complication associated with the treatment of kidney failure with peritoneal dialysis (PD) using a glucose-based dialysate. Mesothelial cells (MCs) take part in the inflammatory processes by producing various cytokines and chemokines, such as monocyte chemoattractant protein 1 (MCP-1) and interleukin 8 (IL-8). The apoptosis of MCs induced by high glucose levels also contributes to complications of PD. High mobility group protein B1 (HMGB1) is an inflammatory factor that has repeatedly been proven to be related to the occurrence of peritoneal dysfunction. Aim: In this study, we aimed to explore the effect and underlying mechanism of endogenous HMGB1 in high-glucose-induced MC injury. Methods: The human peritoneal MC line, HMrSV5 was cultured in high-glucose medium and incubated with recombinant HMGB1. Cellular expression of HMGB1 was blocked using HMGB1 small interfering RNA (siRNA). Apoptosis and production of inflammatory factors as well as the potential intermediary signaling pathways were examined. Results: The major findings of these analyses were: (1) MCs secreted HMGB1 from the nucleus during exposure to high glucose levels; HMGB1 acted in an autocrine fashion on the MCs to promote the production of MCP-1 and IL-8; (2) HMGB1 had little effect on high-glucose-induced apoptosis of the MCs; and (3) HMGB1-mediated MCP-1 and IL-8 production depended on the activation of MAPK signaling pathways. In conclusion, endogenous HMGB1 plays an important role in the inflammatory reaction induced by high glucose on MCs via mitogen-activated protein kinase (MAPK) signaling pathways, but it seems to have little effect on high-glucose-induced apoptosis.
Collapse
Affiliation(s)
- Yuening Chu
- Department of Nephrology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Wang
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Zhihuang Zheng
- Department of Nephrology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuli Lin
- Department of Immunology and Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Rui He
- Department of Immunology and Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jun Liu
- Department of Nephrology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xuguang Yang
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
39
|
Ke X, Yang D, Liang J, Wang X, Wu S, Wang X, Hu C. Human Endothelial Progenitor Cell-Derived Exosomes Increase Proliferation and Angiogenesis in Cardiac Fibroblasts by Promoting the Mesenchymal-Endothelial Transition and Reducing High Mobility Group Box 1 Protein B1 Expression. DNA Cell Biol 2017; 36:1018-1028. [PMID: 28920705 DOI: 10.1089/dna.2017.3836] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Myocardial fibrosis is a characteristic feature of cardiomyopathies. However, no effective strategies to attenuate cardiac fibrosis are currently available. Late-stage endothelial progenitor cells (EPCs) are precursors of endothelial cells (ECs) that repair the heart through a paracrine mechanism. In the present study, we tested whether EPC-derived exosomes regulate the differentiation of fibroblasts into ECs. We isolated late-stage EPCs from human peripheral blood (PB) and used immunofluorescence and flow cytometry to confirm their identity. Next, we isolated exosomes from the EPCs and characterized their morphology using electron microscopy and confirmed the expression of exosome-specific marker proteins using Western blots. We then investigated the in vitro effects of exosomes on the proliferation and angiogenesis of cardiac fibroblasts (CFs) and on the expression of the mesenchymal-endothelial transition (MEndT)-related genes and the myocardial fibrosis-regulated protein, high mobility group box 1 protein B1 (HMGB1). We found that human PB-EPC-derived exosomes enhanced the proliferation and angiogenesis of CFs in vitro. Furthermore, CFs stimulated with these exosomes showed increased expression of the EC-specific markers, like cluster of differentiation 31 and vascular endothelial growth factor receptor 2, and decreased expression of proteins involved in fibrosis, like alpha-smooth muscle actin, vimentin, collagen I, transforming growth factor-beta, and tumor necrosis factor-alpha. In addition, CFs stimulated with human PB-EPC-derived exosomes, inhibited the expression of HMGB1. Taken together, our study demonstrated that EPC-derived exosomes promote the proliferation and angiogenesis of CFs by inhibiting MEndT and decreasing the expression of HMGB1.
Collapse
Affiliation(s)
- Xiao Ke
- 1 Department of Cardiology, Shenzhen Sun Yat-sen Cardiovascular Hospital , Shenzhen, People's Republic of China
| | - Dahao Yang
- 1 Department of Cardiology, Shenzhen Sun Yat-sen Cardiovascular Hospital , Shenzhen, People's Republic of China
| | - Jiawen Liang
- 2 Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Xing Wang
- 2 Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Shaoyun Wu
- 2 Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Xiaoqing Wang
- 1 Department of Cardiology, Shenzhen Sun Yat-sen Cardiovascular Hospital , Shenzhen, People's Republic of China
| | - Chengheng Hu
- 2 Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| |
Collapse
|