Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Adv Sci (Weinh) ; : e2400952, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-39031880

ABSTRACT

Toxoplasma gondii (T. gondii)-associated polymorphic effector proteins are crucial in parasite development and regulating host anti-T. gondii immune responses. However, the mechanism remains obscure. Here, it is shown that Toxoplasma effector dense granules 4 (GRA4) restricts host IFN-I activation. Infection with Δgra4 mutant T. gondii strain induces stronger IFN-I responses and poses a severe threat to host health. Mechanistically, GRA4 binds to phosphorylated TBK1 to promote TRIM27-catalyzed K48-ubiquitination at Lys251/Lys372 residues, which enhances its recognition by autophagy receptor p62, ultimately leading to TBK1 autophagic degradation. Furthermore, an avirulent Δgra4 strain (ME49Δompdc/gra4) is constructed for tumor immunotherapy due to its ability to enhance IFN-I production. Earlier vaccination with ME49Δompdc/gra4 confers complete host resistance to the tumor compared with the classical ME49Δompdc treatment. Notably, ME49Δompdc/gra4 vaccination induces a specific CD64+MAR-1+CD11b+ dendritic cell subset, thereby enhancing T cell anti-tumor responses. Overall, these findings identify the negative role of T. gondii GRA4 in modulating host IFN-I signaling and suggest that GRA4 can be a potential target for the development of T. gondii vaccines and tumor immunotherapy.

2.
J Cell Mol Med ; 28(9): e18349, 2024 May.
Article in English | MEDLINE | ID: mdl-38686493

ABSTRACT

The pathogenesis of trauma-induced heterotopic ossification (HO) in the tendon remains unclear, posing a challenging hurdle in treatment. Recognizing inflammation as the root cause of HO, anti-inflammatory agents hold promise for its management. Malvidin (MA), possessing anti-inflammatory properties, emerges as a potential agent to impede HO progression. This study aimed to investigate the effect of MA in treating trauma-induced HO and unravel its underlying mechanisms. Herein, the effectiveness of MA in preventing HO formation was assessed through local injection in a rat model. The potential mechanism underlying MA's treatment was investigated in the tendon-resident progenitor cells of tendon-derived stem cells (TDSCs), exploring its pathway in HO formation. The findings demonstrated that MA effectively hindered the osteogenic differentiation of TDSCs by inhibiting the mTORC1 signalling pathway, consequently impeding the progression of trauma-induced HO of Achilles tendon in rats. Specifically, MA facilitated the degradation of Rheb through the K48-linked ubiquitination-proteasome pathway by modulating USP4 and intercepted the interaction between Rheb and the mTORC1 complex, thus inhibiting the mTORC1 signalling pathway. Hence, MA presents itself as a promising candidate for treating trauma-induced HO in the Achilles tendon, acting by targeting Rheb for degradation through the ubiquitin-proteasome pathway.


Subject(s)
Ossification, Heterotopic , Proteasome Endopeptidase Complex , Ras Homolog Enriched in Brain Protein , Signal Transduction , Ubiquitin , Animals , Rats , Proteasome Endopeptidase Complex/metabolism , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/etiology , Ossification, Heterotopic/pathology , Signal Transduction/drug effects , Ras Homolog Enriched in Brain Protein/metabolism , Ubiquitin/metabolism , Male , Osteogenesis/drug effects , Tendons/metabolism , Tendons/pathology , Rats, Sprague-Dawley , Tendon Injuries/metabolism , Tendon Injuries/pathology , Tendon Injuries/complications , Proteolysis/drug effects , Cell Differentiation/drug effects , Achilles Tendon/metabolism , Achilles Tendon/pathology , Achilles Tendon/injuries , Disease Models, Animal , Ubiquitination , Mechanistic Target of Rapamycin Complex 1/metabolism , Stem Cells/metabolism , Stem Cells/drug effects
3.
Phytomedicine ; 124: 155323, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38194842

ABSTRACT

BACKGROUND: Currently, there are no specific drugs or targets available for the treatment of tendinopathy. However, inflammation has recently been found to play a pivotal role in tendinopathy progression, thereby identifying it as a potential therapeutic target. Carpaine (CA) exhibits potential anti-inflammatory pharmacological properties and may offer a therapeutic option for tendinopathy. PURPOSE: This study aimed to investigate the effectiveness of CA in addressing tendinopathy and uncovering its underlying mechanisms. METHODS: Herein, the efficacy of CA by local administration in vivo in comparison to the first-line drug indomethacin was evaluated in a mouse collagenase-induced tendinopathy (CIT) model. Furthermore, IL-1ß induced a simulated pathological inflammatory microenvironment in tenocytes to investigate its underlying mechanisms in vitro. Further confirmation experiments were performed by overexpressing or knocking down the selective targets of CA in vivo. RESULTS: The findings demonstrated that CA was dose-dependent in treating tendinopathy and that the high-dose group outperformed the first-line drug indomethacin. Mechanistically, CA selectively bound to and enhanced the activity of the E3 ubiquitin ligase LRSAM1 in tendinopathy. This effect mediated the ubiquitination of p65 at lysine 93, subsequently promoting its proteasomal degradation. As a result, the NF-κB pathway was inactivated, leading to a reduction in inflammation of tendinopathy. Consequently, CA effectively mitigated the progression of tendinopathy. Moreover, the LRSAM1 overexpression demonstrated effectiveness in mitigating the tendinopathy progression and its knockdown abolished the therapeutic effects of CA. CONCLUSION: CA attenuates the progression of tendinopathy by promoting the ubiquitin-proteasomal degradation of p65 via increasing the enzyme activity of LRSAM1. The exploration of LRSAM1 has also unveiled a new potential target for treating tendinopathy based on the ubiquitin-proteasomal pathway.


Subject(s)
Alkaloids , Tendinopathy , Ubiquitin-Protein Ligases , Animals , Mice , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Inflammation/metabolism , Indomethacin , Tendinopathy/drug therapy
4.
Autophagy ; 20(1): 76-93, 2024 01.
Article in English | MEDLINE | ID: mdl-37647255

ABSTRACT

Macroautophagy/autophagy plays an important role in regulating cellular homeostasis and influences the pathogenesis of degenerative diseases. Tendinopathy is characterized by tendon degeneration and inflammation. However, little is known about the role of selective autophagy in tendinopathy. Here, we find that pristimerin (PM), a quinone methide triterpenoid, is more effective in treating tendinopathy than the first-line drug indomethacin. PM inhibits the AIM2 inflammasome and alleviates inflammation during tendinopathy by promoting the autophagic degradation of AIM2 through a PYCARD/ASC-dependent manner. A mechanistic study shows that PM enhances the K63-linked ubiquitin chains of PYCARD/ASC at K158/161, which serves as a recognition signal for SQSTM1/p62-mediated autophagic degradation of the AIM2-PYCARD/ASC complex. We further identify that PM binds the Cys53 site of deubiquitinase USP50 through the Michael-acceptor and blocks the binding of USP50 to PYCARD/ASC, thereby reducing USP50-mediated cleavage of K63-linked ubiquitin chains of PYCARD/ASC. Finally, PM treatment in vivo generates an effect comparable to inflammasome deficiency in alleviating tendinopathy. Taken together, these findings demonstrate that PM alleviates the progression of tendinopathy by modulating AIM2-PYCARD/ASC stability via SQSTM1/p62-mediated selective autophagic degradation, thus providing a promising autophagy-based therapeutic for tendinopathy.Abbreviations: 3-MA: 3-methyladenine; AIM2: absent in melanoma 2; AT: Achilles tenotomy; ATP: adenosine triphosphate; BMDMs: bone marrow-derived macrophages; CHX: cycloheximide; Col3a1: collagen, type III, alpha 1; CQ: chloroquine; Cys: cysteine; DARTS: drug affinity responsive target stability; DTT: dithiothreitol; DUB: deubiquitinase; gDNA: genomic DNA; GSH: glutathione; His: histidine; IL1B/IL-1ß: interleukin 1 beta; IND: indomethacin; IP: immunoprecipitation; LPS: lipopolysaccharide; MMP: mitochondrial membrane potential; NLRP3: NLR family, pyrin domain containing 3; PM: pristimerin; PYCARD/ASC: PYD and CARD domain containing; SN: supernatants; SOX9: SRY (sex determining region Y)-box 9; SQSTM1: sequestosome 1; Tgfb: transforming growth factor, beta; TIMP3: tissue inhibitor of metalloproteinase 3; TNMD: tenomodulin; TRAF6: TNF receptor-associated factor 6; Ub: ubiquitin; USP50: ubiquitin specific peptidase 50; WCL: whole cell lysates.


Subject(s)
Inflammasomes , Tendinopathy , Humans , Inflammasomes/metabolism , Sequestosome-1 Protein/metabolism , Autophagy/genetics , Macroautophagy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammation , Ubiquitin/metabolism , Indomethacin/pharmacology , Deubiquitinating Enzymes/metabolism , Interleukin-1beta/metabolism , DNA-Binding Proteins/metabolism
5.
Cytotechnology ; 75(6): 461-472, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37841958

ABSTRACT

The NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 4-like 2 (NDUFA4L2) gene has been reported to be upregulated in colorectal cancer (CRC) and is associated with worse prognosis. However, the specific function and underlying mechanism of NDUFA4L2 in colon adenocarcinoma (COAD) under hypoxia has never been investigated. Our study discovered that hypoxia promoted the viability, metastasis, and epithelial-mesenchymal transition (EMT) of COAD cells. Besides, hypoxia-induced HIF-1α upregulated the expression of NDUFA4L2 which served as an oncogene and an independent diagnostic and prognostic marker in COAD. Under hypoxic environment, NDUFA4L2 mediated the viability, metastasis, and epithelial-EMT of COAD cells. Additionally, the ROS-dependent PI3K/Akt signaling was activated by NDUFA4L2 in COAD in hypoxia and NDUFA4L2 facilitated the malignant behaviors of hypoxia-treated COAD cells by elevating ROS production. Collectively, abundant NDUFA4L2 expression induced by HIF-1α under hypoxia promoted the development of COAD through activation of the PI3K/AKT signaling in a ROS-dependent manner, indicating NDUFA4L2 as a promising target in COAD diagnosis and treatment.

6.
mBio ; 14(4): e0351222, 2023 Aug 31.
Article in English | MEDLINE | ID: mdl-37366613

ABSTRACT

Stringent control of the type I interferon (IFN-I) signaling is critical for host immune defense against infectious diseases, yet the molecular mechanisms that regulate this pathway remain elusive. Here, we show that Src homology 2 containing inositol phosphatase 1 (SHIP1) suppresses IFN-I signaling by promoting IRF3 degradation during malaria infection. Genetic ablation of Ship1 in mice leads to high levels of IFN-I and confers resistance to Plasmodium yoelii nigeriensis (P.y.) N67 infection. Mechanistically, SHIP1 promotes the selective autophagic degradation of IRF3 by enhancing K63-linked ubiquitination of IRF3 at lysine 313, which serves as a recognition signal for NDP52-mediated selective autophagic degradation. In addition, SHIP1 is downregulated by IFN-I-induced miR-155-5p upon P.y. N67 infection and severs as a feedback loop of the signaling crosstalk. This study reveals a regulatory mechanism between IFN-I signaling and autophagy, and verifies SHIP1 can be a potential target for therapeutic intervention against malaria and other infectious diseases. IMPORTANCE Malaria remains a serious disease affecting millions of people worldwide. Malaria parasite infection triggers tightly controlled type I interferon (IFN-I) signaling that plays a critical role in host innate immunity; however, the molecular mechanisms underlying the immune responses are still elusive. Here, we discover a host gene [Src homology 2-containing inositol phosphatase 1 (SHIP1)] that can regulate IFN-I signaling by modulating NDP52-mediated selective autophagic degradation of IRF3 and significantly affect parasitemia and resistance of Plasmodium-infected mice. This study identifies SHIP1 as a potential target for immunotherapies in malaria and highlights the crosstalk between IFN-I signaling and autophagy in preventing related infectious diseases. SHIP1 functions as a negative regulator during malaria infection by targeting IRF3 for autophagic degradation.

7.
Sci Adv ; 9(25): eadg2339, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37352355

ABSTRACT

Stringent control of type I interferon (IFN-I) signaling is critical to potent innate immune responses against viral infection, yet the underlying molecular mechanisms are still elusive. Here, we found that Van Gogh-like 2 (VANGL2) acts as an IFN-inducible negative feedback regulator to suppress IFN-I signaling during vesicular stomatitis virus (VSV) infection. Mechanistically, VANGL2 interacted with TBK1 and promoted the selective autophagic degradation of TBK1 via K48-linked polyubiquitination at Lys372 by the E3 ligase TRIP, which serves as a recognition signal for the cargo receptor OPTN. Furthermore, myeloid-specific deletion of VANGL2 in mice showed enhanced IFN-I production against VSV infection and improved survival. In general, these findings revealed a negative feedback loop of IFN-I signaling through the VANGL2-TRIP-TBK1-OPTN axis and highlighted the cross-talk between IFN-I and autophagy in preventing viral infection. VANGL2 could be a potential clinical therapeutic target for viral infectious diseases, including COVID-19.


Subject(s)
Interferon Type I , Protein Serine-Threonine Kinases , Virus Diseases , Animals , Mice , Autophagy , Cell Polarity , Protein Serine-Threonine Kinases/metabolism , Transcription Factors , Virus Diseases/immunology , Interferon Type I/immunology
8.
J Oncol ; 2023: 8538240, 2023.
Article in English | MEDLINE | ID: mdl-36778919

ABSTRACT

Background: The relationship between H. pylori infection and gastric cancer (GC) has been widely studied, and H. pylori is considered as the main factor. Utilizing bioinformatics analysis, this study examined gene signatures related to progressing H. pylori-associated GC. Materials and Methods: The dataset GSE13195 was chosen to search for abnormally expressed genes in H. pylori-associated GC and normal tissues. The TCGA-STAD database was chosen to verify the expression of key genes in GC and normal tissues. Results: In GSE13195, a total of 332 differential expression genes (DEGs) were screened. The results of weighted gene co-expression network analysis showed that the light cyan, plum2, black, and magenta4 modules were associated with stages (T3, T2, and T4), while the orangered4, salmon2, pink, and navajowhite2 modules were correlated with lymph node metastasis (N3, N2, and N0). Based on the results of DEGs and hub genes, a total of 7 key genes (ADAM28, FCER1G, MRPL14, SOSTDC1, TYROBP, C1QC, and C3) were screened out. These gene mRNA levels were able to distinguish between normal and H. pylori-associated GC tissue using receiver operating characteristic curves. After transcriptional level verification and survival analysis, ADAM28 and C1QC were excluded. An immune infiltration study revealed that key genes were involved in regulating the infiltration levels of cells associated with innate immune response, antigen presentation process, humoral immune response, or Tcell-mediated immune response. In addition, drugs targeting FCER1G and TYROBP have been approved and are under investigation. Conclusion: Our study identified five key genes involved in H. pylori-associated GC tumorigenesis. Patients with higher levels of C3 expression had a poorer prognosis than those with lower levels. In addition, these key genes may serve as biomarkers and therapeutic targets for H. pylori-associated GC diagnosis, targeted therapy, and immunotherapy in the future.

9.
mBio ; 13(6): e0236122, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36214572

ABSTRACT

Innate immunity acts as the first line of defense against pathogen invasion. During Toxoplasma gondii infection, multiple innate immune sensors are activated by invading microbes or pathogen-associated molecular patterns (PAMPs). However, how inflammasome is activated and its regulatory mechanisms during T. gondii infection remain elusive. Here, we showed that the infection of PRU, a lethal type II T. gondii strain, activates inflammasome at the early stage of infection. PRU tachyzoites, RNA and soluble tachyzoite antigen (STAg) mainly triggered the NLRP3 inflammasome, while PRU genomic DNA (gDNA) specially activated the AIM2 inflammasome. Furthermore, mice deficient in AIM2, NLRP3, or caspase-1/11 were more susceptible to T. gondii PRU infection, and the ablation of inflammasome signaling impaired antitoxoplasmosis immune responses by enhancing type I interferon (IFN-I) production. Blockage of IFN-I receptor fulfilled inflammasome-deficient mice competent immune responses as WT mice. Moreover, we have identified that the suppressor of cytokine signaling 1 (SOCS1) is a key negative regulator induced by inflammasome-activated IL-1ß signaling and inhibits IFN-I production by targeting interferon regulatory factor 3 (IRF3). In general, our study defines a novel protective role of inflammasome activation during toxoplasmosis and identifies a critical regulatory mechanism of the cross talk between inflammasome and IFN-I signaling for understanding infectious diseases. IMPORTANCE As a key component of innate immunity, inflammasome is critical for host antitoxoplasmosis immunity, but the underlying mechanisms are still elusive. In this study, we found that inflammasome signaling was activated by PAMPs of T. gondii, which generated a protective immunity against T. gondii invasion by suppressing type I interferon (IFN-I) production. Mechanically, inflammasome-coupled IL-1ß signaling triggered the expression of negative regulator SOCS1, which bound to IRF3 to inhibit IFN-I production. The role of IFN-I in anti-T. gondii immunity is little studied and controversial, and here we also found IFN-I is harmful to host antitoxoplasmosis immunity by using knockout mice and recombinant proteins. In general, our study identifies a protective role of inflammasomes to the host during T. gondii infection and a novel mechanism by which inflammasome suppresses IFN-I signaling in antitoxoplasmosis immunity, which will likely provide new insights into therapeutic targets for toxoplasmosis and highlight the cross talk between innate immune signaling in infectious diseases prevention.


Subject(s)
Communicable Diseases , Interferon Type I , Toxoplasma , Toxoplasmosis , Animals , Mice , Inflammasomes , Toxoplasma/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pathogen-Associated Molecular Pattern Molecules , Immunity, Innate , Mice, Knockout
10.
J Oncol ; 2022: 5137627, 2022.
Article in English | MEDLINE | ID: mdl-35794986

ABSTRACT

Gastric cancer (GC) is one of the most common malignancies, and novel prognostic biomarkers for it are urgently required. This study is aimed at screening a group of immune-related lncRNAs (IRLs) in predicting the prognosis of GC patients. Genetic and clinical information from the 360 GC patients was included in this study. Eight IRLs in lncRNA-miRNA-mRNA network were screened out according to differential expression analysis. A novel risk score model with three IRLs (MIR4435-1HG, UCA1, and RP11-617F23.1) were identified, and patients were assigned to a high-risk group and a low-risk group. Patients in the low-risk group had a better prognosis. In addition, two nomograms were developed to predict the prognosis of GC. We evaluated the correlation between IRLs and the immune infiltration level of GC using TIMER. Furthermore, we verified that RP11-617F23.1 was significantly upregulated in human GC tissues compared with their adjacent tissues. And, patients with high RP11-617F23.1 expression in tumor tissues had poorer survival. In conclusion, we established a novel risk model based on IRLs for predicting the prognosis of GC. Meanwhile, a novel IRL, RP11-617F23.1, could serve as a predictor of prognosis for patients with GC.

11.
Int Immunopharmacol ; 110: 108962, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35753124

ABSTRACT

Intervertebral disc degeneration (IDD) is a common orthopedic multifactorial disease associated with spine-related disorders, such as low back pain. Recent studies have shown that both platelet-rich plasma (PRP) and exosomes could be used to treat IDD, but the effects and mechanism of PRP-derived exosomes in the treatment of IDD are still unclear. This study showed that PRP-derived exosomes inhibited the polarization of M1 macrophages by regulating the NF-κB and MAPK pathways and affected the polarization of M2 macrophages by regulating STAT6 phosphorylation. Additionally, PRP-derived exosomes promoted the autophagic degradation of NLRP3 by increasing NLRP3 ubiquitination and reducing IL-1ß and Caspase-1 production. Moreover, PRP-derived exosomes could reduce IL-1ß-induced apoptosis of nucleus pulposus cells. Lastly, in vivo experiments confirmed that PRP-derived exosomes reduced the expression of inflammatory mediators and apoptotic factors, which could thereby alleviate the progression of IDD. Taken together, these data showed that PRP-derived exosomes could alleviate the IDD-associated inflammation by regulating the ubiquitination and autophagic degradation of NLRP3 inflammasome, providing new insights into the treatment of IDD.


Subject(s)
Exosomes , Intervertebral Disc Degeneration , Intervertebral Disc , Platelet-Rich Plasma , Exosomes/metabolism , Humans , Intervertebral Disc/metabolism , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/therapy , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Platelet-Rich Plasma/metabolism
12.
J Cell Mol Med ; 26(12): 3483-3494, 2022 06.
Article in English | MEDLINE | ID: mdl-35582962

ABSTRACT

Tendinopathy is mainly characterized by local pain, functional limitation and decreased athletic ability, which seriously affects the quality of life of patients and the career of athletes. Farrerol (FA), one of the main active compounds extracted from Rhododendron and plants in the Rhododendron family, has a wide range of pharmacological activities, such as immunomodulatory, anti-inflammatory and antiviral effects. However, the effect of FA on tendinopathy is unclear. Here, we investigated the pharmacological effect and mechanism of FA in tendon injury through collagenase-induced tendinopathy in vivo and RSL3-induced tenocytes injury in vitro. The results showed that FA alleviated the infiltration of inflammatory cells, promoted tenogenesis and improved mechanical properties of the Achilles tendon in rats. In addition, ferroptosis inducer RSL3 inhibits the tenogenesis in vitro and in vivo, which accelerates the progression of tendinopathy. Moreover, FA effectively inhibited iron accumulation and alleviated ferroptosis in the Achilles tendon. Using in vitro experiments, we found that FA antagonized ferroptosis by reducing lipid peroxidation and iron accumulation in tenocytes. Finally, we found that glutathione peroxidase 4 silencing could block the protective effect of FA on ferroptosis of tenocytes. Therefore, the results of this study suggest that FA can relieve collagenase-induced tendinopathy by inhibiting ferroptosis, and reveal that FA may be a potentially effective drug for the treatment of tendinopathy in the future.


Subject(s)
Chromones , Ferroptosis , Tendinopathy , Animals , Chromones/pharmacology , Collagenases/administration & dosage , Ferroptosis/drug effects , Humans , Iron/metabolism , Quality of Life , Rats , Tendinopathy/chemically induced , Tendinopathy/drug therapy , Tendinopathy/metabolism
13.
Nutrients ; 14(8)2022 Apr 18.
Article in English | MEDLINE | ID: mdl-35458235

ABSTRACT

With the development of an aging population, tendinopathy has become a common musculoskeletal disease in the elderly with a high recurrence rate and no curative treatment. The inflammation mediated by NF-κB signaling plays an important role in tendon senescence and degeneration. Friedelin (FR) is a triterpenoid derived from green plants, which has a variety of pharmacological functions, such as analgesia, anti-inflammation, antioxidation, and anti-tumor functions. However, the role and mechanism of FR in tendinopathy are unclear. Here, we found that FR improved the mechanical strength of the Achilles tendon, restored the orderly arrangement of collagen fibers, reduced inflammatory cell infiltration, and promoted tenogenesis, thereby blocking the progression of tendinopathy. Mechanistically, FR promoted the autophagic degradation of p65 by enhancing the interaction between p62 and p65 and effectively inhibited the activation of the NF-κB pathway, thus alleviating the inflammatory response of tenocytes. In addition, FR recruited E3 ubiquitin enzyme RNF182 to increase the K48-linked ubiquitination of p65 and promoted p62-mediated autophagic degradation. Furthermore, blocking ubiquitination reversed the degradation of p65 by FR. Therefore, these findings identify the new pharmacological mechanism of the anti-inflammatory effect of FR and provide a new candidate drug for the treatment of tendinopathy.


Subject(s)
Tendinopathy , Triterpenes , Animals , Collagenases , Mice , NF-kappa B/metabolism , Tendinopathy/chemically induced , Tendinopathy/drug therapy , Triterpenes/pharmacology , Triterpenes/therapeutic use
14.
J Cell Mol Med ; 25(23): 10825-10836, 2021 12.
Article in English | MEDLINE | ID: mdl-34783166

ABSTRACT

Osteoporosis is characterized by increased bone fragility, and the drugs used at present to treat osteoporosis can cause adverse reactions. Gentiopicroside (GEN), a class of natural compounds with numerous biological activities such as anti-resorptive properties and protective effects against bone loss. Therefore, the aim of this work was to explore the effect of GEN on bone mesenchymal stem cells (BMSCs) osteogenesis for a potential osteoporosis therapy. In vitro, BMSCs were exposed to GEN at different doses for 2 weeks, whereas in vivo, ovariectomized osteoporosis was established in mice and the therapeutic effect of GEN was evaluated for 3 months. Our results in vitro showed that GEN promoted the activity of alkaline phosphatase, increased the calcified nodules in BMSCs and up-regulated the osteogenic factors (Runx2, OSX, OCN, OPN and BMP2). In vivo, GEN promoted the expression of Runx2, OCN and BMP2, increased the level of osteogenic parameters, and accelerated the osteogenesis of BMSCs by activating the BMP pathway and Wnt/ß-catenin pathway, effect that was inhibited using the BMP inhibitor Noggin and Wnt/ß-catenin inhibitor DKK1. Silencing the ß-catenin gene and BMP2 gene blocked the osteogenic differentiation induced by GEN in BMSCs. This block was also observed when only ß-catenin was silenced, although the knockout of BMP2 did not affect ß-catenin expression induced by GEN. Therefore, GEN promotes BMSC osteogenesis by regulating ß-catenin-BMP signalling, providing a novel strategy in the treatment of osteoporosis.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Iridoid Glucosides/pharmacology , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , beta Catenin/metabolism , Animals , Bone and Bones/drug effects , Bone and Bones/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Female , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Osteoporosis/metabolism , Recombinant Proteins/metabolism , Up-Regulation/drug effects , Wnt Signaling Pathway/drug effects
15.
J Gastrointest Oncol ; 12(3): 1164-1179, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34295565

ABSTRACT

BACKGROUND: Pancreatic adenocarcinoma (PAAD) has a high rate of mortality. Unfortunately, it is difficult to diagnosis. This study aimed to develop a more in-depth understanding of the disease. METHODS: A total of 177 patients with PAAD were recruited from The Cancer Genome Atlas (TCGA) database. Microarray analysis was performed to identify differentially expressed genes (DEGs) in PAAD. The microarray data were adapted to the ingenuity pathway analysis (IPA) for annotation and visualization, followed by protein-protein interaction (PPI) network analysis. In vitro transwell migration assays were conducted to explore the molecular and functional characteristics of pancreatic adenocarcinoma cells (PANC-1) with stable low expression of G-protein signaling modulator 2 (GPSM2). Expression of GPSM2 and the associated hub genes were detected by reverse transcription-quantitative polymerase chain reaction (qPCR). RESULTS: The overexpression of GPSM2 was proved in PAAD, as compared with the healthy tissues, as well as its correlation with history of chronic pancreatitis, T stage, TNM stage and tumor grade. We described it as an independent prognostic factor and found that it could influence the infiltration of immune cells in the tumor microenvironment. Silencing of GPSM2 restrained the and migration of the cells. Microarray analysis identified 1,631 DEGs in PAAD cells. The PPI network analysis identified hub genes including CD44, ITGB1, ITGB5, ITGA2, ITGA5, AKT1, EGFR, NRAS and MAP2K1, and their relationship with GPSM2 was confirmed by qPCR. CONCLUSIONS: GPSM2 is a novel prognostic factor and therapeutic target for PAAD. GPSM2 promoted the migration of pancreatic adenocarcinoma cells .Targeting GPSM2 and its downstream genes may prolong the survival time of patients with PAAD.

16.
J Cell Mol Med ; 25(14): 6664-6678, 2021 07.
Article in English | MEDLINE | ID: mdl-34117708

ABSTRACT

Synovial macrophage polarization and inflammation are essential for osteoarthritis (OA) development, yet the molecular mechanisms and regulation responsible for the pathogenesis are still poorly understood. Here, we report that pseudolaric acid B (PAB) attenuated articular cartilage degeneration and synovitis during OA. PAB, a diterpene acid, specifically inhibited NF-κB signalling and reduced the production of pro-inflammatory cytokines, which further decreased M1 polarization and vessel formation. We further provide in vivo and in vitro evidences that PAB suppressed NF-κB signalling by stabilizing PPARγ. Using PPARγ antagonist could abolish anti-inflammatory effect of PAB and rescue the activation of NF-κB signalling during OA. Our findings identify a previously unrecognized role of PAB in the regulation of OA and provide mechanisms by which PAB regulates NF-κB signalling through PPARγ, which further suggest targeting synovial inflammation or inhibiting vessel formation at early stage could be an effective preventive strategy for OA.


Subject(s)
Diterpenes/pharmacology , Osteoarthritis/drug therapy , PPAR gamma/genetics , Synovitis/drug therapy , Animals , Blood Vessels/drug effects , Blood Vessels/growth & development , Cartilage, Articular/drug effects , Cartilage, Articular/growth & development , Cartilage, Articular/pathology , Chondrocytes/drug effects , Disease Models, Animal , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/parasitology , Mice , NF-kappa B/genetics , Osteoarthritis/genetics , Osteoarthritis/pathology , RAW 264.7 Cells , Signal Transduction/drug effects , Synovitis/genetics , Synovitis/pathology , Transcription Factor RelA/genetics
17.
J Cell Biochem ; 120(9): 16025-16036, 2019 09.
Article in English | MEDLINE | ID: mdl-31074104

ABSTRACT

This paper aims to probe into the effect of sweroside (SOS) in osteoporosis (OP) and explains mechanisms of its molecular. Applying the ovariectomized (OVX) mouse model investigates the preventive effect of SOS against postmenopausal OP after 3 months of SOS treatment (120 mg/kg/day). Using hematoxylin and eosin (HE) staining and micro computed tomography (CT) observed the morphology of OP in each group. Immunohistochemical staining (IHC) was used to examine osteoblast markers. Experiments in vitro, bone marrow mesenchymal stem cells (BMSCs) from C57/BL6 mice were treated with SOS for 14 days. The staining of alizarin red and alkaline phosphatase activity were measured, and the presentation of osteoblast markers was detected by quantitative reverse transcription PCR. BMSCs were also treated with 1 µg/mL SOS with or without rapamycin, the expression of protein S6 (PS6), P-mTOR, runt-related transcription factor 2 (RUNX2), OSX, and osteocalcin (OCN) was detected by Western blotting. Experiments in vivo, HE results show that SOS can alleviate OP, CT results show that there are lower trabecular thickness, bone mineral density, and trabecular number in control OVX mice than those in the OVX + SOS group. IHC results showed that SOS can promote the expression of osteogenic markers and immunofluorescent results show that SOS can promote mTORC1 signal activation. Experiments in vitro revealed that SOS stimulated the activation of the mTORC1 signaling pathway and upregulated RUNX2, OSX, and OCN, rapamycin can reverse it. Our findings demonstrated that differentiated BMSCs into osteoblasts can be promoted by SOS via upregulating the expression of P-mTOR, PS6, RUNX2, OSX, and OCN. SOS effectively prevented OP by hyperactivation of the mTORC1/PS6 signaling pathway.


Subject(s)
Iridoid Glucosides/administration & dosage , Mechanistic Target of Rapamycin Complex 1/metabolism , Mesenchymal Stem Cells/cytology , Osteoporosis, Postmenopausal/drug therapy , Animals , Biomarkers/metabolism , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Disease Models, Animal , Drug Administration Schedule , Female , Humans , Iridoid Glucosides/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Osteocalcin/genetics , Osteocalcin/metabolism , Osteoporosis, Postmenopausal/diagnostic imaging , Osteoporosis, Postmenopausal/genetics , Osteoporosis, Postmenopausal/metabolism , Signal Transduction/drug effects , Sp7 Transcription Factor/genetics , Sp7 Transcription Factor/metabolism , X-Ray Microtomography
18.
Inflammation ; 42(2): 496-505, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30315524

ABSTRACT

Sweroside (SW), as a bioactive herbal ingredient, has anti-inflammatory effects. Protective effects of SW on IL-1ß-stimulated articular chondrocytes, however, has not been fully understood. This study was to explore the anti-inflammatory effects and further to investigate the possible mechanism underlying SW effect on IL-1ß-stimulated rat articular chondrocytes. Rat articular chondrocytes were cultured with or without SW for 1 h, and then stimulated with IL-1ß for 24 h. ELISA analysis was used to measure the production of NO and PGE2. Western blot was to detect the expression of iNOS and COX-2. Furthermore, the mRNA expression of MMP-1, MMP3, MMP13, and ADAMTS-5 were measured by q-PCR. These results demonstrated that SW significantly inhibited IL-1ß-induced NO and PGE2 production, as well as MMP-1, MMP3, MMP13, and ADAMTS-5 mRNA expression. Moreover, SW also suppressed IL-1ß-induced NF-κB activation and iκ-B degradation, S6K1 and S6 phosphorylation. In conclusion, these results strongly demonstrated that the anti-inflammatory activity of SW is in part mediated by suppressing NF-κB and mTORC1 signaling, which was expected to be a promising drug target of osteoarthritis therapy.


Subject(s)
Chondrocytes/drug effects , Inflammation/drug therapy , Interleukin-1beta/adverse effects , Iridoid Glucosides/pharmacology , Signal Transduction/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Cartilage, Articular , Dinoprostone/biosynthesis , Inflammation/chemically induced , Iridoid Glucosides/therapeutic use , Mechanistic Target of Rapamycin Complex 1/metabolism , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Plants, Medicinal/chemistry , Protective Agents/pharmacology , Rats
19.
Cell Physiol Biochem ; 48(1): 293-303, 2018.
Article in English | MEDLINE | ID: mdl-30016763

ABSTRACT

BACKGROUND/AIMS: Narrowing of the lumbar spinal canal is a condition called lumbar spinal stenosis (LSS) and is a high-morbidity problem in the elderly. LSS is commonly caused by hypertrophy of the ligamentum flavum (HLF). Previous studies showed that fibrosis of the ligamentum flavum (LF) largely contributed to HLF. However, the underlying pathomechanism remains unclear. Insulin-like growth factor-1 (IGF-1) is known to have an intimate relationship with fibrosis in various tissues. Nevertheless, currently, there are few studies regarding IGF-1 in HLF. In this study, we investigated the role of IGF-1 in HLF and its potential molecular mechanism of action. METHODS: First, the IGF-1, phosphorylation of IGF-1 receptor (pIGF-1R), phosphorylation of AKT (pAKT), phosphorylation of S6(pS6), collagen I and collagen III expression levels were examined via immunohistochemistry and Western blotting in LF tissues from patients with LSS or Non-LSS. Second, primary LF cells were isolated from adults with a normal LF thickness and were cultured with different concentrations of IGF-1 with or without NVP-AEW541/rapamycin. RESULTS: The results showed that IGF-1, pIGF-1R, pAKT, pS6, collagen I and collagen III protein expression in the LSS group was significantly higher than that in the Non-LSS group. Meanwhile, pIGF-1R, pAKT, pS6, collagen I and collagen III protein expression was significantly enhanced in LF cells after IGF-1 exposure, which can be notably blocked by NVP-AEW541. In addition, pS6, collagen I and collagen III protein expression was blocked by rapamycin. CONCLUSIONS: Enhanced IGF-1 promotes the synthesis of collagen I and collagen III via the mTORC1 signaling pathway, which eventually contributes to hypertrophy of the ligamentum flavum.


Subject(s)
Hypertrophy/pathology , Insulin-Like Growth Factor I/metabolism , Ligamentum Flavum/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Signal Transduction , Aged , Case-Control Studies , Cell Survival , Collagen Type I/metabolism , Collagen Type III/metabolism , Female , Gene Expression/drug effects , Humans , Ligamentum Flavum/cytology , Ligamentum Flavum/diagnostic imaging , Male , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Middle Aged , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology
20.
Biomed Res Int ; 2018: 3849760, 2018.
Article in English | MEDLINE | ID: mdl-29693006

ABSTRACT

BACKGROUND: Tendon-bone healing is a reconstructive procedure which requires a tendon graft healing to a bone tunnel or to the surface of bone after the junction injury between tendon, ligament, and bone. The surgical reattachment of tendon to bone often fails due to regeneration failure of the specialized tendon-bone junction. MATERIALS AND METHODS: An extra-articular tendon-bone healing rat model was established to discuss the effect of the baicalein 10 mg/(kg·d) in accelerating tendon-bone healing progress. Also, tendon-derived stem cells (TDSCs) were treated with various concentrations of baicalein or dickkopf-1 (DKK-1) to stimulate differentiation for 14 days. RESULTS: In vivo, tendon-bone healing strength of experiment group was obviously stronger than the control group in 3 weeks as well as in 6 weeks. And there were more mature fibroblasts, more Sharpey fibers, and larger new bone formation area treated intragastrically with baicalein compared with rats that were treated with vehicle for 3 weeks and 6 weeks. In vitro, after induction for 14 days, the expressions of osteoblast differentiation markers, that is, alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2), osteocalcin (OCN), osterix (OSX), and collagen I, were upregulated and Wnt/ß-catenin signaling pathway was enhanced in TDSCs. The effect of DKK-1 significantly reduced the effect of baicalein on the osteogenic differentiation. CONCLUSION: These data suggest that baicalein may stimulate TDSCs osteogenic differentiation via activation of Wnt/ß-catenin signaling pathway to accelerate tendon-bone healing.


Subject(s)
Bone and Bones/drug effects , Flavanones/pharmacology , Tendons/drug effects , Wnt Signaling Pathway/drug effects , Wound Healing/drug effects , beta Catenin/metabolism , Alkaline Phosphatase/metabolism , Animals , Bone and Bones/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteocalcin/drug effects , Osteocalcin/metabolism , Osteogenesis/drug effects , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects , Stem Cells/metabolism , Tendons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL