Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
1.
Cell Mol Life Sci ; 81(1): 236, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38795203

ABSTRACT

Chemoresistance is the main obstacle in the clinical treatment of osteosarcoma (OS). In this study, we investigated the role of EF-hand domain-containing protein 1 (EFHD1) in OS chemotherapy resistance. We found that the expression of EFHD1 was highly correlated with the clinical outcome after chemotherapy. We overexpressed EFHD1 in 143B cells and found that it increased their resistance to cell death after drug treatment. Conversely, knockdown of EFHD1 in 143BR cells (a cisplatin-less-sensitive OS cell line derived from 143B cells) increased their sensitivity to treatment. Mechanistically, EFHD1 bound to adenine nucleotide translocase-3 (ANT3) and inhibited its conformational change, thereby inhibiting the opening of the mitochondrial membrane permeability transition pore (mPTP). This effect could maintain mitochondrial function, thereby favoring OS cell survival. The ANT3 conformational inhibitor carboxyatractyloside (CATR), which can promote mPTP opening, enhanced the chemosensitivity of EFHD1-overexpressing cells when combined with cisplatin. The ANT3 conformational inhibitor bongkrekic acid (BKA), which can inhibit mPTP opening, restored the resistance of EFHD1 knockdown cells. In conclusion, our results suggest that EFHD1-ANT3-mPTP might be a promising target for OS therapy in the future.


Subject(s)
Cell Proliferation , Cisplatin , Drug Resistance, Neoplasm , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Osteosarcoma , Humans , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Mitochondrial Permeability Transition Pore/metabolism , Drug Resistance, Neoplasm/drug effects , Cell Proliferation/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Cell Line, Tumor , Cisplatin/pharmacology , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Adenine Nucleotide Translocator 3/metabolism , Adenine Nucleotide Translocator 3/genetics , Antineoplastic Agents/pharmacology , Mitochondria/metabolism , Mitochondria/drug effects , Animals , Mice , Protein Binding
2.
J Biol Chem ; 299(6): 104823, 2023 06.
Article in English | MEDLINE | ID: mdl-37187293

ABSTRACT

An imbalance of human mesenchymal stem cells (MSCs) adipogenic and osteogenic differentiation plays an important role in the pathogenesis of osteoporosis. Our previous study verified that Adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1 (APPL1)/myoferlin deficiency promotes adipogenic differentiation of MSCs by blocking autophagic flux in osteoporosis. However, the function of APPL1 in the osteogenic differentiation of MSCs remains unclear. This study aimed to investigate the role of APPL1 in the osteogenic differentiation of MSCs in osteoporosis and the underlying regulatory mechanism. In this study, we demonstrated the downregulation of APPL1 expression in patients with osteoporosis and osteoporosis mice. The severity of clinical osteoporosis was negatively correlated with the expression of APPL1 in bone marrow MSCs. We found that APPL1 positively regulates the osteogenic differentiation of MSCs in vitro and in vivo. Moreover, RNA sequencing showed that the expression of MGP, an osteocalcin/matrix Gla family member, was significantly upregulated after APPL1 knockdown. Mechanistically, our study showed that reduced APPL1 impaired the osteogenic differentiation of mesenchymal stem cells by facilitating Matrix Gla protein expression to disrupt the BMP2 pathway in osteoporosis. We also evaluated the significance of APPL1 in promoting osteogenesis in a mouse model of osteoporosis. These results suggest that APPL1 may be an important target for the diagnosis and treatment of osteoporosis.


Subject(s)
Adaptor Proteins, Signal Transducing , Calcium-Binding Proteins , Mesenchymal Stem Cells , Osteoporosis , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Cells, Cultured , Membrane Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Muscle Proteins/metabolism , Osteogenesis , Osteoporosis/metabolism , Calcium-Binding Proteins/metabolism , Matrix Gla Protein
3.
Cell Mol Life Sci ; 80(11): 325, 2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37831180

ABSTRACT

Increasing evidence indicates that circular RNAs (circRNAs) accumulate in aging tissues and nonproliferating cells due to their high stability. However, whether upregulation of circRNA expression mediates stem cell senescence and whether circRNAs can be targeted to alleviate aging-related disorders remain unclear. Here, RNA sequencing analysis of differentially expressed circRNAs in long-term-cultured mesenchymal stem cells (MSCs) revealed that circSERPINE2 expression was significantly increased in late passages. CircSERPINE2 small interfering RNA delayed MSC senescence and rejuvenated MSCs, while circSERPINE2 overexpression had the opposite effect. RNA pulldown followed by mass spectrometry revealed an interaction between circSERPINE2 and YBX3. CircSERPINE2 increased the affinity of YBX3 for ZO-1 through the CCAUC motif, resulting in the sequestration of YBX3 in the cytoplasm, inhibiting the association of YBX3 with the PCNA promoter and eventually affecting p21 ubiquitin-mediated degradation. In addition, our results demonstrated that senescence-related downregulation of EIF4A3 gave rise to circSERPINE2. In vivo, intra-articular injection of si-circSerpine2 restrained native joint-resident MSC senescence and cartilage degeneration in mice with aging-related osteoarthritis. Taken together, our findings provide strong evidence for a regulatory role for the circSERPINE2/YBX3/PCNA/p21 axis in MSC senescence and the therapeutic potential of si-circSERPINE2 in alleviating aging-associated syndromes, such as osteoarthritis.


Subject(s)
Mesenchymal Stem Cells , Osteoarthritis , Mice , Animals , Proliferating Cell Nuclear Antigen , RNA, Circular/genetics , RNA, Circular/metabolism , Mesenchymal Stem Cells/metabolism , Cellular Senescence/genetics , RNA, Small Interfering/metabolism , Osteoarthritis/metabolism
4.
Int J Mol Sci ; 25(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38892028

ABSTRACT

Amino acid permeases (AAPs) transporters are crucial for the long-distance transport of amino acids in plants, from source to sink. While Arabidopsis and rice have been extensively studied, research on foxtail millet is limited. This study identified two transcripts of SiAAP9, both of which were induced by NO3- and showed similar expression patterns. The overexpression of SiAAP9L and SiAAP9S in Arabidopsis inhibited plant growth and seed size, although SiAAP9 was found to transport more amino acids into seeds. Furthermore, SiAAP9-OX transgenic Arabidopsis showed increased tolerance to high concentrations of glutamate (Glu) and histidine (His). The high overexpression level of SiAAP9 suggested its protein was not only located on the plasma membrane but potentially on other organelles, as well. Interestingly, sequence deletion reduced SiAAP9's sensitivity to Brefeldin A (BFA), and SiAAP9 had ectopic localization on the endoplasmic reticulum (ER). Protoplast amino acid uptake experiments indicated that SiAAP9 enhanced Glu transport into foxtail millet cells. Overall, the two transcripts of SiAAP9 have similar functions, but SiAAP9L shows a higher colocalization with BFA compartments compared to SiAAP9S. Our research identifies a potential candidate gene for enhancing the nutritional quality of foxtail millet through breeding.


Subject(s)
Arabidopsis , Endoplasmic Reticulum , Gene Expression Regulation, Plant , Plant Proteins , Plants, Genetically Modified , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/growth & development , Plant Proteins/genetics , Plant Proteins/metabolism , Endoplasmic Reticulum/metabolism , Seeds/metabolism , Seeds/genetics , Seeds/growth & development , Setaria Plant/genetics , Setaria Plant/metabolism , Setaria Plant/growth & development , Amino Acid Transport Systems/metabolism , Amino Acid Transport Systems/genetics , Protein Transport , Brefeldin A/pharmacology , Amino Acids/metabolism , Glutamic Acid/metabolism
5.
Stem Cells ; 40(5): 508-522, 2022 05 27.
Article in English | MEDLINE | ID: mdl-35403694

ABSTRACT

Bone marrow (BM) adipose tissue (BMAT), a unique adipose depot, plays an important role in diseases such as osteoporosis and bone metastasis. Precise control of mesenchymal stem cell (MSC) differentiation is critical for BMAT formation and regeneration. Here, we show that death associated protein kinase 1 (DAPK1) negatively regulates BM adipogenesis in vitro and in vivo. Prx1creDapk1loxp/loxp mice showed more adipocytes in the femur than Dapk1loxp/loxp mice. Further mechanistic analyses revealed that DAPK1 inhibits p38 mitogen-activated protein kinase (MAPK) signaling in the nucleus by binding the p38 isoform MAPK14, decreasing p38 nuclear activity, which subsequently inhibits BM adipogenesis. The inhibitory effect of DAPK1 against MAPK14 was independent of its kinase activity. In addition, the decreased DAPK1 was observed in the BM-MSCs of ageing mice. Our results reveal a previously undescribed function for DAPK1 in the regulation of adipogenesis and may also reveal the underlying mechanism of BMAT formation in ageing.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Mesenchymal Stem Cells , Mitogen-Activated Protein Kinase 14 , Adipogenesis , Animals , Bone Marrow , Bone Marrow Cells , Cell Differentiation , Death-Associated Protein Kinases/genetics , Death-Associated Protein Kinases/metabolism , Death-Associated Protein Kinases/pharmacology , Mesenchymal Stem Cells/metabolism , Mice , Mitogen-Activated Protein Kinase 14/metabolism , Protein Isoforms/metabolism
6.
J Nanobiotechnology ; 21(1): 168, 2023 May 26.
Article in English | MEDLINE | ID: mdl-37231465

ABSTRACT

Ankylosing spondylitis (AS) is a common rheumatic disorder distinguished by chronic inflammation and heterotopic ossification at local entheses sites. Currently available medications, including nonsteroidal anti-inflammatory drugs (NSAIDs), disease-modifying anti-rheumatic drugs (DMARDs) and TNF inhibitors, are limited by side effects, high costs and unclear inhibitory effects on heterotopic ossification. Herein, we developed manganese ferrite nanoparticles modified by the aptamer CH6 (CH6-MF NPs) that can efficiently scavenge ROS and actively deliver siRNA into hMSCs and osteoblasts in vivo for effective AS treatment. CH6-MF NPs loaded with BMP2 siRNA (CH6-MF-Si NPs) effectively suppressed abnormal osteogenic differentiation under inflammatory conditions in vitro. During their circulation and passive accumulation in inflamed joints in the Zap70mut mouse model, CH6-MF-Si NPs attenuated local inflammation and rescued heterotopic ossification in the entheses. Thus, CH6-MF NPs may be an effective inflammation reliever and osteoblast-specific delivery system, and CH6-MF-Si NPs have potential for the dual treatment of chronic inflammation and heterotopic ossification in AS.


Subject(s)
Ossification, Heterotopic , Spondylitis, Ankylosing , Mice , Animals , Spondylitis, Ankylosing/drug therapy , Spondylitis, Ankylosing/pathology , Osteogenesis , Inflammation/drug therapy , Inflammation/pathology , Osteoblasts , RNA, Small Interfering/pharmacology , Ossification, Heterotopic/pathology
7.
J Nanobiotechnology ; 21(1): 280, 2023 Aug 19.
Article in English | MEDLINE | ID: mdl-37598147

ABSTRACT

Sustained inflammatory invasion leads to joint damage and progressive disability in several autoimmune rheumatic diseases. In recent decades, targeting M1 macrophage polarization has been suggested as a promising therapeutic strategy for autoimmune arthritis. P300/CBP-associated factor (PCAF) is a histone acetyltransferase (HAT) that exhibits a strong positive relationship with the proinflammatory microenvironment. However, whether PCAF mediates M1 macrophage polarization remains poorly studied, and whether targeting PCAF can protect against autoimmune arthritis in vivo remains unclear. Commonly used drugs can cause serious side effects in patients because of their extensive and nonspecific distribution in the human body. One strategy for overcoming this challenge is to develop drug nanocarriers that target the drug to desirable regions and reduce the fraction of drug that reaches undesirable targets. In this study, we demonstrated that PCAF inhibition could effectively inhibit M1 polarization and alleviate arthritis in mice with collagen-induced arthritis (CIA) via synergistic NF-κB and H3K9Ac blockade. We further designed dextran sulfate (DS)-based nanoparticles (DSNPs) carrying garcinol (a PCAF inhibitor) to specifically target M1 macrophages in inflamed joints of the CIA mouse model via SR-A-SR-A ligand interactions. Compared to free garcinol, garcinol-loaded DSNPs selectively targeted M1 macrophages in inflamed joints and significantly improved therapeutic efficacy in vivo. In summary, our study indicates that targeted PCAF inhibition with nanoparticles might be a promising strategy for treating autoimmune arthritis via M1 macrophage polarization inhibition.


Subject(s)
Arthritis , NF-kappa B , Humans , Animals , Mice , Terpenes , Macrophages
8.
Cell Mol Life Sci ; 79(9): 488, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35984564

ABSTRACT

An imbalance of human mesenchymal stem cells (hMSCs) adipogenic and osteogenic differentiation is crucial in the pathogenesis of osteoporosis, and elucidation of the underlying mechanism is urgently needed. APPL1, an adaptor protein of the adiponectin receptor, was recently shown to be closely related to bone mass. However, the role of APPL1 in the imbalance of hMSC differentiation in osteoporosis is unclear. Therefore, we aimed to explore the mechanisms by which APPL1 alters hMSCs adipogenic differentiation in osteoporosis. Here, we found that APPL1 expression was downregulated in elderly patients with osteoporosis and in mouse osteoporosis model. APPL1 negatively regulated hMSC adipogenic differentiation in vivo and in vitro. Mechanistically, by enhancing ubiquitination-mediated Myoferlin degradation, downregulated APPL1 expression increased the risk of lysosome dysfunction during hMSCs adipogenic differentiation. Lysosomal dysfunction inhibited autophagy flux by suppressing autophagosome degradation and promoted hMSC differentiation towards the adipocyte lineage. Our findings suggest that APPL1/Myoferlin downregulation promoted hMSCs adipogenic differentiation by inhibiting autophagy flux, further impairing the balance of hMSCs adipogenic and osteogenic differentiation in osteoporosis; the APPL1/ Myoferlin axis may be a promising diagnostic and therapeutic target for osteoporosis.


Subject(s)
Adaptor Proteins, Signal Transducing , Membrane Proteins , Mesenchymal Stem Cells , Muscle Proteins , Osteoporosis , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adipogenesis/genetics , Aged , Animals , Autophagy/physiology , Calcium-Binding Proteins , Cell Differentiation/physiology , Cells, Cultured , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Muscle Proteins/metabolism , Osteogenesis/genetics , Osteoporosis/genetics , Osteoporosis/metabolism
9.
Acta Biochim Biophys Sin (Shanghai) ; 55(8): 1275-1287, 2023 Jun 26.
Article in English | MEDLINE | ID: mdl-37365870

ABSTRACT

Glucocorticoid-induced osteoporosis (GIOP), one of the most common and serious adverse effects associated with glucocorticoid administration, manifests as decreased bone formation and increased bone resorption, eventually culminating in bone loss. Galangin (GAL) is a flavonoid extracted from the medicinal herbal galangal that possesses a variety of pharmacological activities and can inhibit osteoclastogenesis. However, the effects of GAL on GIOP remain unclear. Our study aims to explore the effects of GAL on GIOP in mice and the underlying mechanism. Our results show that GAL markedly mitigates the severity of dexamethasone (Dex)-induced osteoporosis in mice and potentiates osteogenic differentiation in mouse bone marrow-derived mesenchymal stem cells (BMSCs). Furthermore, GAL also significantly counteracts Dex-mediated suppression of osteogenic differentiation and autophagy in human BMSCs. GAL augments PKA/CREB-mediated autophagic flux in BMSCs and the bones of osteoporotic mice. GAL-mediated osteogenic differentiation in Dex-treated BMSCs is significantly decreased by the PKA inhibitor H89 and autophagy inhibitor 3-methyladenine. Collectively, our data indicate that GAL can ameliorate GIOP, partly by augmenting the mineralization of BMSCs by potentiating PKA/CREB-mediated autophagic flux, highlighting its potential therapeutic use in treating glucocorticoid-related osteoporosis.


Subject(s)
Glucocorticoids , Osteoporosis , Humans , Mice , Animals , Glucocorticoids/adverse effects , Osteogenesis , Osteoporosis/chemically induced , Osteoporosis/drug therapy , Osteoporosis/metabolism , Flavonoids/pharmacology , Signal Transduction , Cell Differentiation , Autophagy
10.
Phytother Res ; 37(1): 252-270, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36104214

ABSTRACT

Osteoporosis, a systemic bone disease that is characterized by a reduction in bone mass and destruction of bone microstructure, is becoming a serious problem worldwide. Bone marrow mesenchymal stem cells (BMSCs) can differentiate into bone-forming osteoblasts, and play an important role in maintaining homeostasis of bone metabolism, thus being a potential therapeutic target for osteoporosis. Although the phytochemical alpinetin (APT) has been reported to possess a variety of pharmacological activities, it is still unclear whether APT can influence the osteogenic differentiation of on BMSCs and if it can improve osteoporosis. In this study, we found that APT treatment was able to enhance osteogenic differentiation levels of human BMSCs in vitro and mouse ones in vivo as revealed by multiple osteogenic markers including increased alkaline phosphatase activity and osteocalcin expression. Mechanistically, the protein kinase A (PKA)/mTOR/ULK1 signaling was involved in the action of APT to enhance the osteogenic differentiation of BMSCs. In addition, oral administration of APT significantly mitigated the bone loss in a dexamethasone-induced mouse model of osteoporosis through strengthening PKA signaling and autophagy. Altogether, these data demonstrate that APT promotes osteogenic differentiation in BMSCs by augmenting the PKA/mTOR/ULK1 autophagy signaling, highlighting its potential therapeutic application for treating osteoporotic diseases.


Subject(s)
Mesenchymal Stem Cells , Osteoporosis , Mice , Humans , Animals , Osteogenesis , Osteoporosis/drug therapy , Cell Differentiation , Mesenchymal Stem Cells/metabolism , TOR Serine-Threonine Kinases/metabolism , Autophagy , Cells, Cultured , Bone Marrow Cells/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/therapeutic use
11.
Ren Fail ; 45(2): 2256414, 2023.
Article in English | MEDLINE | ID: mdl-37724523

ABSTRACT

Introduction: We explored the relationship and the predictive value of serum fibroblast growth factor 21 (FGF21) with all-cause mortality, major adverse cardiovascular events (MACEs) and pneumonia in hemodialysis (HD) patients.Methods: A total of 388 Chinese HD patients from two HD centers were finally enrolled in this prospective cohort study (registration number: ChiCTR 1900028249) between January 2018 and December 2018. Serum FGF21 was detected. Patients were followed up with a median period of 47 months to record the MACEs and pneumonia until death or 31 December 2022.Results: The incidence of all-cause mortality, MACEs and pneumonia in HD patients were 20.6%, 29.6%, and 34.8%, respectively. The optimal cutoffs for FGF21 to predict all-cause mortality, MACEs and pneumonia were 437.57 pg/mL, 216.99 pg/mL and 112.79 pg/mL. Multivariate Cox regression analyses showed that FGF21, as a categorical variable, was an independent predictor for all-cause mortality, MACEs and pneumonia (HR, 3.357, 95% CI, 2.128-5.295, p < 0.001; HR, 1.575, 95% CI, 1.046-2.371, p = 0.029; HR, 1.784; 95% CI, 1.124-2.830; p = 0.014, respectively). The survival nomogram, MACEs-free survival nomogram and pneumonia-free survival nomogram based on FGF21 constructed for individualized assessment of HD patients had a high C-index with 0.841, 0.706 and 0.734.Conclusion: Higher serum FGF21 is an independent predictor of all-cause mortality, MACEs and pneumonia in HD patients.


Subject(s)
Fibroblast Growth Factors , Renal Dialysis , Humans , Fibroblast Growth Factors/blood , Prospective Studies , Renal Dialysis/adverse effects , East Asian People
12.
J Stroke Cerebrovasc Dis ; 32(4): 107055, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36801498

ABSTRACT

OBJECTIVES: Time from onset to reperfusion affects mortality and favorable outcomes in patients with acute ischemic stroke (AIS). To evaluate effects of a real-time feedback mobile application on critical time intervals and functional outcomes in stroke emergency management. METHODS: We recruited patients with clinically suspected acute stroke from December 1st, 2020 until July 30st, 2022. All Patients had a non-contrast computed tomography (CT) and were included only if they had AIS. We divided the patients into two groups based on the date of availability on mobile application: pre-APP group and post-APP group. Onset to Door time (ODT), Door to Imaging Time (DIT), Door to Needle Time (DNT), Door to Puncture Time (DPT), Door to Recanalization Time (DRT), National Institutes of Health Stroke Scale (NIHSS) and modified Rankin Scale (mRS) were compared between two groups. RESULTS: We retrospectively enrolled 312 AIS patients who were assigned into the pre-APP group (n = 159) and post-APP group (n = 153). The median ODT time and median admission NIHSS score were not significantly different between the two groups at baseline assessment. The median (IQR) DIT [44 (30-60) min vs 28 (20-36) min, P < 0.01] and DNT [44 (36.25-52) min vs 39 (29-45) min, P = 0.02] both decreased significantly in two groups. However, median DPT and DRT time showed no significant differences. The proportion of mRS score of 0 to 2 at day 90 was significantly higher in the post-App group than in the pre-App group, at 82.4% and 71.7%, respectively (dominance ratio OR=1.84, 95% CI: 1.07 to 3.16, P = 0.03). CONCLUSION: The present findings indicate that the real-time feedback of stroke emergency management used by a mobile application have potential for shortening the DIT and DNT time and improve the prognosis of stroke patients.


Subject(s)
Ischemic Stroke , Mobile Applications , Stroke , Humans , Ischemic Stroke/diagnostic imaging , Ischemic Stroke/therapy , Retrospective Studies , Feedback , Stroke/diagnostic imaging , Stroke/therapy , Tomography, X-Ray Computed , Treatment Outcome , Thrombolytic Therapy/adverse effects , Time-to-Treatment , Fibrinolytic Agents/adverse effects
13.
Stem Cells ; 39(5): 581-599, 2021 05.
Article in English | MEDLINE | ID: mdl-33547700

ABSTRACT

Dysregulated angiogenesis of mesenchymal stem cells (MSCs) is closely related to inflammation and disrupted bone metabolism in patients with various autoimmune diseases. However, the role of MSCs in the development of abnormal angiogenesis in patients with ankylosing spondylitis (AS) remains unclear. In this study, we cultured human umbilical vein endothelial cells (HUVECs) with bone marrow-derived MSCs from patients with AS (ASMSCs) or healthy donors (HDMSCs) in vitro. Then, the cocultured HUVECs were assayed using a cell counting kit-8 (CCK-8) to evaluate the cell proliferation. A wound healing assay was performed to investigate cell migration, and a tube formation assay was conducted to determine the angiogenesis efficiency. ASMSCs exhibited increased angiogenesis, and increased expression of SMAD-specific E3 ubiquitin ligase 2 (Smurf2) in MSCs was the main cause of abnormal angiogenesis in patients with AS. Downregulation of Smurf2 in ASMSCs blocked angiogenesis, whereas overexpression of Smurf2 in HDMSCs promoted angiogenesis. The pro-angiogenic effect of Smurf2 was confirmed by the results of a Matrigel plug assay in vivo. By functioning as an E3 ubiquitin ligase in MSCs, Smurf2 regulated the levels of pentraxin 3 (PTX3), which has been shown to suppress angiogenesis through the PTX3-fibroblast growth factor 2 pathway. Moreover, Smurf2 transcription was regulated by activating transcription factor 4-induced endoplasmic reticulum stress. In conclusion, these results identify novel roles of Smurf2 in negatively regulating PTX3 stability and promoting angiogenesis in ASMSCs.


Subject(s)
C-Reactive Protein/genetics , Neovascularization, Pathologic/genetics , Serum Amyloid P-Component/genetics , Spondylitis, Ankylosing/genetics , Ubiquitin-Protein Ligases/genetics , Activating Transcription Factor 4/genetics , Cell Movement/genetics , Coculture Techniques , Endoplasmic Reticulum Stress/genetics , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation, Developmental/genetics , Healthy Volunteers , Human Umbilical Vein Endothelial Cells , Humans , Mesenchymal Stem Cells/metabolism , Neovascularization, Pathologic/complications , Neovascularization, Pathologic/pathology , Spondylitis, Ankylosing/complications , Spondylitis, Ankylosing/pathology , Ubiquitin-Protein Ligases/antagonists & inhibitors
14.
J Biomed Sci ; 29(1): 73, 2022 Sep 21.
Article in English | MEDLINE | ID: mdl-36127734

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) exhibit two bidirectional immunomodulatory abilities: proinflammatory and anti-inflammatory regulatory effects. Long noncoding RNAs (lncRNAs) have important functions in the immune system. Previously, we performed high-throughput sequencing comparing lncRNA expression profiles between MSCs cocultured with or without CD14+ monocytes and screened out a new lncRNA termed lncRNA MCP1 regulatory factor (MRF). However, the mechanism of MRF in MSCs is still unknown. METHODS: MRF expression was quantified via qRT-PCR. RNA interference and lentiviruses were used to regulate MRF expression. The immunomodulatory effects of MSCs on monocytes were evaluated via monocyte migration and macrophage polarization assays. RNA pull-down and mass spectrometry were utilized to identify downstream factors of MRF. A dual-luciferase reporter assay was applied to analyze the transcription factors regulating MRF. qRT-PCR, western blotting and ELISAs were used to assess MCP1 expression. A human monocyte adoptive transfer mouse model was applied to verify the function of MRF in vivo. RESULTS: MRF was upregulated in MSCs during coculture with CD14+ monocytes. MRF increased monocyte recruitment by upregulating the expression of monocyte chemotactic protein (MCP1). Knockdown of MRF enhanced the regulatory effect of MSCs on restraining M1 polarization and facilitating M2 polarization. Mechanistically, MRF bound to the downstream protein heterogeneous nuclear ribonucleoprotein D (HNRNPD) to upregulate MCP1 expression, and the transcription factor interferon regulatory factor 1 (IRF1) activated MRF transcription early during coculture. The human monocyte adoptive transfer model showed that MRF downregulation in MSCs inhibited monocyte chemotaxis and enhanced the effects of MSCs to inhibit M1 macrophage polarization and promote M2 polarization in vivo. CONCLUSION: We identified the new lncRNA MRF, which exhibits proinflammatory characteristics. MRF regulates the ability of MSCs to accelerate monocyte recruitment and modulate macrophage polarization through the HNRNPD-MCP1 axis and initiates the proinflammatory regulatory process in MSCs, suggesting that MRF is a potential target to improve the clinical effect of MSC-based therapy or correct MSC-related immunomodulatory dysfunction under pathological conditions.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D , Mesenchymal Stem Cells , RNA, Long Noncoding , Animals , Anti-Inflammatory Agents/pharmacology , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Heterogeneous-Nuclear Ribonucleoprotein D/pharmacology , Humans , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/pharmacology , Mesenchymal Stem Cells/metabolism , Mice , Monocyte Chemoattractant Proteins/metabolism , Monocyte Chemoattractant Proteins/pharmacology , Monocytes/metabolism , RNA, Long Noncoding/metabolism
15.
Blood Purif ; 51(3): 280-287, 2022.
Article in English | MEDLINE | ID: mdl-34638121

ABSTRACT

INTRODUCTION: The impact of early-onset peritonitis (EOP) on patients with diabetes undergoing peritoneal dialysis (PD) has not been adequately addressed. We therefore sought to investigate the effects of EOP on the therapeutic response to management and long-term prognostic outcomes in patients with diabetes undergoing PD. METHODS: For this retrospective cohort study, we analyzed the data for patients with end-stage renal disease, who were also suffering from diabetes mellitus and had undergone PD between January 1, 2013, and December 31, 2018. EOP was defined as the first episode of peritoneal dialysis-related peritonitis (PDAP) occurring within 12 months of PD initiation. All patients were divided into an EOP group and a later-onset peritonitis (LOP) group. Clinical data, treatment results, and outcomes were compared between groups. RESULTS: Ultimately, 202 patients were enrolled for the analysis. Compared to the EOP group, the LOP group had more Streptococcus (p = 0.033) and Pseudomonas (p = 0.048). Patients with diabetes in the EOP group were less likely to have PDAP-related death (OR 0.13, CI: 0.02-0.82, p = 0.030). Patients with diabetes in the EOP group were more likely to have multiple episodes of PDAP and had higher rates of technical failure and poorer patient survival than those in the LOP group, as indicated by Kaplan-Meier analysis (p = 0.019, p = 0.004, and p < 0.001). In the multivariate Cox proportional hazards model, EOP was a significant predictor for multiple PDAP (HR 4.20, CI: 1.48-11.96, p = 0.007), technical failure (HR 6.37, CI: 2.21-18.38, p = 0.001), and poorer patient survival (HR 3.09, CI: 1.45-6.58, p = 0.003). CONCLUSIONS: The occurrence of EOP is significantly associated with lower rates of PDAP-related death and poorer clinical outcomes in patients with diabetes undergoing PD.


Subject(s)
Diabetes Mellitus , Kidney Failure, Chronic , Peritoneal Dialysis , Peritonitis , Female , Humans , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/therapy , Male , Peritoneal Dialysis/adverse effects , Peritoneal Dialysis/methods , Peritonitis/drug therapy , Peritonitis/therapy , Retrospective Studies , Risk Factors
16.
Sensors (Basel) ; 22(16)2022 Aug 11.
Article in English | MEDLINE | ID: mdl-36015754

ABSTRACT

To overcome the difficulty of collecting the working resistance and working trajectory of a wheel loader, this paper constructs a statics model of the bucket working resistance and a kinematics model of the working trajectory in the shoveling process and analyzes the key parameters of measuring the working resistance and working trajectory. Based on this, a working resistance and working trajectory acquisition strategy is proposed. To verify the effectiveness of the acquisition strategy, the in-service operation data of fine sand and loose soil shoveled by the wheel loader are collected and analyzed. Then, the test-fitted working resistance and working trajectory are obtained, and the working trajectory is input into the RecurDyn-EDEM co-simulation model to obtain the simulation-fitted working resistance. Considering the complex working conditions of the wheel loader, it is difficult to obtain accurate working resistance, and the actual working resistance is also a relative value. Therefore, a strong correlation between the two curves indicates that the acquisition strategy of the wheel loader's working trajectory and working resistance proposed in this paper is feasible.


Subject(s)
Soil , Computer Simulation
17.
Drug Dev Res ; 83(1): 97-104, 2022 02.
Article in English | MEDLINE | ID: mdl-34173680

ABSTRACT

Chronic unpredictable mild stress (CUMS) can cause a series of depressive symptoms in depression patients. Recently, notoginsenoside R1 (NGR1) has been reported to play crucial roles in the anti-inflammatory, antioxidant, and anti-apoptotic. However, the role and mechanisms of NGR1 in improving symptoms of depressive behavior remain unknown. Evaluating and identifying its value and exploring the mechanisms of NGR1 on CUMS-induced depressive behavior were the aims of this study. Here, rats were separated into five different groups and treated with or without different concentrations of the NGR1. Then, the body weight, sucrose preference rate, immobility time, crossing number, rearing number, and grooming number were determined to evaluate the effect of NGR1 on improving the depressive behavior of CUMS rats. Subsequently, the morphology of hippocampal neurons and protein expression of brain-derived neurotrophic factor in each group were examined by hematoxylin and eosin staining and western blot to show the neuroprotective effects of NGR1. Furthermore, the mRNA and protein expression of TNF-α, IL-6, and IL-1ß were also detected by quantitative polymerase chain reaction (qPCR) and enzyme-linked immunosorbent assay to verify the anti-inflammatory effects of NGR1 on CUMS rats. In addition, the cell apoptosis-related proteins were examined to reveal that NGR1 can inhibit cell apoptosis in CUMS rats. Moreover, it was confirmed that NGR1 attenuated the symptoms of depressive behavior by mediated PI3K/Akt/NF-κB pathway. Together, this study shows that NGR1 improves depressive behavior induced by chronic stress in rats through activation of PI3K/AKT/NF-κB pathway.


Subject(s)
Ginsenosides , NF-kappa B , Animals , Depression/drug therapy , Depression/etiology , Depression/metabolism , Disease Models, Animal , Ginsenosides/pharmacology , Ginsenosides/therapeutic use , Hippocampus/metabolism , Humans , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Stress, Psychological/complications , Stress, Psychological/drug therapy
18.
J Stroke Cerebrovasc Dis ; 31(2): 106222, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34839235

ABSTRACT

OBJECTIVE: A self-rating post stroke depression scale (PSDS) showed a good reliability and validity to assess severity of depressive symptoms among stroke patients. This study aimed to retest the psychometric properties of PSDS in different types of post-stroke depression (PSD). MATERIALS AND METHODS: A total of 170 stroke patients were recruited in the study. 82 and 25 patients were respectively diagnosed as PSD symptoms disorder (PSDSD) and PSD disorder (PSDD) patients according to their respective diagnostic criteria. The PSDS and the 9-item Patient Health Questionnaire (PHQ-9) were used to assess the severity of depression. Cronbach α, Spearman rank coefficient and independent sample t-test were conducted to examine reliability, internal consistency and discriminate validity. Then the receiver operating characteristic curve and Youden index were used to performance evaluation and cut-off value respectively in different subtypes of PSD patients. RESULTS: The Cronbach α of PSDS was 0.857, indicting a good reliability. The Spearman correlation coefficient between PSDS and PHQ-9 was 0.942 (P<0.001). The discriminate validity displayed significant difference between PSDSD as well as PSDD and no depression patients (all P<0.001). 5/24 and 10/24 were the cut-off value for PSDSD and PSDD patients. CONCLUSIONS: PSDS is a useful screen tool with an acceptable psychometric properties for estimation of different subtypes of PSD patients.


Subject(s)
Depression , Patient Health Questionnaire , Stroke , Depression/diagnosis , Depression/etiology , Humans , Psychometrics , Reproducibility of Results , Stroke/complications
19.
Mol Med ; 27(1): 20, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33637046

ABSTRACT

BACKGROUND: Transplantation of bone marrow-derived mesenchymal stem cells (BM-MSCs) embedded in a bio-compatible matrix has been demonstrated as a promising strategy for the treatment of bone defects. This study was designed to explore the effect and mechanism of exosomes derived from mature dendritic cells (mDC-Exo) on the BM-MSCs-mediated bone regeneration using the matrix support in an athymic rat model of femoral bone defect. METHODS: The BM-MSCs were isolated from rats and incubated with osteoblast induction medium, exosomes derived from immature DCs (imDC-Exo), mDC-Exo, and miR-335-deficient mDC-Exo. BM-MSCs treated without or with mDC-Exo were embedded in a bio-compatible matrix (Orthoss®) and then implanted into the femoral bone defect of athymic rats. RESULTS: mDC-Exo promoted the proliferation and osteogenic differentiation of BM-MSCs by transferring miR-335. Mechanistically, exosomal miR-335 inhibited Hippo signaling by targeting large tongue suppressor kinase 1 (LATS1) and thus promoted the proliferation and osteogenic differentiation of BM-MSCs. Animal experiments showed that mDC-Exo enhanced BM-MSCs-mediated bone regeneration after bone defect, and this effect was abrogated when miR-335 expression was inhibited in mDC-Exo. CONCLUSION: mDC-Exo promoted osteogenic differentiation of BM-MSCs and enhanced BM-MSCs-mediated bone regeneration after femoral bone defect in athymic rats by transferring miR-335.


Subject(s)
Bone Regeneration , Dendritic Cells/cytology , Exosomes , Mesenchymal Stem Cell Transplantation , MicroRNAs , Animals , Bone Diseases/diagnostic imaging , Bone Diseases/genetics , Bone Diseases/surgery , Cells, Cultured , Coculture Techniques , Exosomes/genetics , Exosomes/metabolism , Female , Femur/diagnostic imaging , Femur/immunology , Femur/injuries , Femur/surgery , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats, Nude , X-Ray Microtomography
20.
Cancer Cell Int ; 21(1): 648, 2021 Dec 04.
Article in English | MEDLINE | ID: mdl-34863167

ABSTRACT

BACKGROUND: Osteosarcoma (OS) patients with lung metastasis have poor prognoses, and effective therapeutic strategies for delaying or inhibiting the spread of lung metastasis from the primary OS site are lacking. Hence, it is critical to elucidate the underlying mechanisms of OS metastasis and to identify additional new effective treatment strategies for patients. METHODS: Differential expression and functional analyses were performed to identify key genes and relevant signaling pathways associated with OS lung metastasis. The expression of CCR9 in OS cell lines and tissues was measured by RT-qPCR, western blotting and immunohistochemistry. Cell migration and invasion were assessed by wound healing and Transwell Matrigel invasion assays, respectively. The regulatory relationship between CCR9 and the Wnt/ß-catenin signaling pathway was further evaluated by rescue experiments. RESULTS: The expression of CCR9 was elevated in OS cell lines and patients with lung metastasis. CCR9 promoted MG63 and HOS cell migration and invasion by activating the Wnt/ß-catenin signaling pathway. Furthermore, knockdown of CCR9 repressed epithelial-mesenchymal transition (EMT) by downregulating mesenchymal markers (N-cadherin and Vimentin) and EMT-associated transcription factors (twist and snail) and upregulating an epithelial marker (E-cadherin). CONCLUSIONS: Our findings suggest that CCR9 promotes EMT by activating Wnt/ß-catenin pathways to promote OS metastasis. CCR9 may be a promising therapeutic target to inhibit lung metastasis and serve as a novel prognostic marker for OS.

SELECTION OF CITATIONS
SEARCH DETAIL