Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Front Genet ; 14: 1073138, 2023.
Article in English | MEDLINE | ID: mdl-37007943

ABSTRACT

The plant homeodomain (PHD) finger refers to a protein motif that plays a key role in the recognition and translation of histone modification marks by promoting gene transcriptional activation and silencing. As an important member of the PHD family, the plant homeodomain finger protein 14 (PHF14) affects the biological behavior of cells as a regulatory factor. Several emerging studies have demonstrated that PHF14 expression is closely associated with the development of some cancers, but there is still no feasible pan-cancer analysis. Based on existing datasets from the Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO), we performed a systematic analysis of the oncogenic role of the PHF14 gene in 33 human cancers. The expression level of PHF14 was significantly different between different types of tumors and adjacent normal tissues, and the expression or genetic alteration of PHF14 gene was closely related to the prognosis of most cancer patients. Levels of cancer-associated fibroblasts (CAFs) infiltration in various cancer types were also observed to correlate with PHF14 expression. In some tumors, PFH14 may play a role in tumor immunity by regulating the expression levels of immune checkpoint genes. In addition, the results of enrichment analysis showed that the main biological activities of PHF14 were related to various signaling pathways or chromatin complex effects. In conclusion, our pan-cancer research shows that the expression level of PHF14 is closely related to the carcinogenesis and prognosis of certain tumors, which needs to be further verified by more experiments and more in-depth mechanism exploration.

2.
Int Immunopharmacol ; 117: 109884, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36805201

ABSTRACT

BACKGROUND: Fractures caused by osteoporosis (OP) are one of the main causes of death in the elderly, bringing a heavy burden to the country and society. The imbalance between osteoblast-mediated osteogenesis and osteoclast-mediated bone resorption is an important cause of OP. Therefore, finding drugs that can regulate this dynamic balance can be an important way to treat osteoporosis. Surfactin is a highly effective biosurfactant derived from Bacillus subtilis and it has been proven to have various pharmacological effects in previous studies, but its effect on bone metabolism remains unknown. Here, we performed a study on the role and mechanism of Surfactin in inhibiting osteoclastogenesis and its possible mechanism as well as the role in promoting osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). METHODS: We investigated the effect of Surfactin on osteoclast differentiation and osteogenic differentiation in vitro and in vivo. The effect of Surfactin on the activity of osteoclastogenesis and osteogenesis was verified by CCK-8 assay, quantitative Real-time polymerase chain reaction (qPCR) and Western blotting analysis were used to verify the effect of Surfactin on osteoclast and osteogenic differentiation-specific genes and proteins. The effect of Surfactin on TRAP、ALP activity and mineral deposition was verified by TRAP、ALP and ARS staining. We then used an ovariectomy-induced osteoporosis mice model to observe the effect of Surfactin in vivo. RESULTS: Surfactin is noncytotoxic to BMMs, RAW264.7, and BMSCs. And it can effectively inhibit osteoclastogenesis and promote osteogenic differentiation. Moreover, we found that Surfactin can inhibit the differentiation of osteoclasts through the NF-κB signaling pathway. Surfactin can also alleviate bone loss in ovariectomy-induced osteoporosis mice. CONCLUSIONS: Our results suggest that Surfactin can inhibit osteoclastogenesis through the NF-κB signaling pathway, promote the osteogenic differentiation of BMSCs, and also can effectively alleviate bone loss in ovariectomy-induced osteoporosis mice.


Subject(s)
Bone Resorption , Osteoporosis , Female , Mice , Animals , Humans , Osteogenesis , NF-kappa B/metabolism , Osteoclasts , Signal Transduction , Bone Resorption/metabolism , Osteoporosis/metabolism , Cell Differentiation , Estrogens/metabolism , RANK Ligand/metabolism , Ovariectomy/adverse effects
3.
Colloids Surf B Biointerfaces ; 222: 113029, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36436402

ABSTRACT

Osteoporosis is an age-related metabolic disease of bone, resulting in bone pain and even bone fragility and brittle fracture. Inhibiting overactive osteoclasts while promoting osteoblast activity is an ideal way to treat osteoporosis. Previous studies have demonstrated that natural compounds, such as curcumin (Cur) have dual roles both in promoting bone formation and inhibiting bone resorption, making them promising candidates for osteoporosis treatment. However, their poor water solubility, high dosage of curative effect and significant toxicity to other organs have largely limited their clinical translations. In this study, a novel method was reported to conjugate Cur and poly(amidoamine) dendrimers (PAD) using hexachlorocyclotriphosphazene (HCCP) as the linkage through a one-pot reaction, forming stable and uniform Cur loaded nanospheres (HCCP-Cur-PAD, HCP NPs). Owing to the hydrophilicity of PAD and hydrophobicity of Cur, HCP NPs can self-assemble into nanoparticles with particle size of 138.8 ± 78.7 nm and display excellent water dispersity. The loading capacity of Cur can reach 27.2% and it can be released from HCP NPs with pH-responsiveness. In vitro experimental results demonstrated that the HCP NPs entered lysosomes by endocytosis and proved dual anti-osteoporosis effects of inhibiting osteoclasts and promoting osteoblasts.


Subject(s)
Curcumin , Dendrimers , Nanoparticles , Solubility , Hydrogen-Ion Concentration , Particle Size , Drug Delivery Systems/methods
4.
Cancer Immunol Immunother ; 72(2): 493-507, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35963895

ABSTRACT

High rates of relapse and poor prognosis confer an urgent need for novel therapeutic agents for B cell non-Hodgkin lymphomas (B-NHLs). Herein, we describe a human IgG-like anti-CD79b/CD3 bispecific antibody (IBI38D9-L) that selectively depletes antigen-positive malignant B cells as an alternative treatment option for relapsed or refractory NHL patients. The antitumor activity and mechanism of action of IBI38D9-L were investigated in vitro using B-NHL cell lines and human primary effector cells and in vivo using xenograft models reconstituted with human PBMCs (peripheral blood mononuclear cells). Pharmacokinetic (PK) properties and preclinical toxicology were evaluated in cynomolgus monkeys and HSC-NPG mice. IBI38D9-L exerted potent B cell killing as well as T cell activation and proliferation in a tumor cell-dependent manner in vitro and was active against B-NHL cell lines with various CD79b expression levels. Subcutaneous xenograft tumors in NOG mice engrafted with human PBMCs were eradicated by IBI38D9-L treatment. Moreover, IBI38D9-L-treated mice showed a strong infiltration of activated T cells. In HSC-NPG mice, IBI38D9-L resulted in potent B cell depletion in peripheral blood and induced only slight body weight loss and cytokine release syndrome without significant toxicological findings. In cynomolgus monkeys, IBI38D9-L was well tolerated with good pharmacokinetic profiles. Collectively, these preclinical efficacy and safety data provide strong scientific rationales for using anti-CD79b/CD3 bispecific antibody as a promising therapeutic agent for B cell malignancies.


Subject(s)
Antibodies, Bispecific , Neoplasms , Humans , Mice , Animals , Macaca fascicularis , Leukocytes, Mononuclear , Antibodies, Bispecific/pharmacology , B-Lymphocytes , Neoplasms/metabolism , CD3 Complex
5.
Cell Rep Med ; 3(6): 100660, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35732156

ABSTRACT

Glucocorticoid-induced tumor necrosis factor receptor (GITR) is a co-stimulatory receptor and an important target for cancer immunotherapy. We herein present a potent FcγR-independent GITR agonist IBI37G5 that can effectively activate effector T cells and synergize with anti-programmed death 1 (PD1) antibody to eradicate established tumors. IBI37G5 depends on both antibody bivalency and GITR homo-dimerization for efficient receptor cross-linking. Functional analyses reveal bell-shaped dose responses due to the unique 2:2 antibody-receptor stoichiometry required for GITR activation. Antibody self-competition is observed after concentration exceeded that of 100% receptor occupancy (RO), which leads to antibody monovalent binding and loss of activity. Retrospective pharmacokinetics/pharmacodynamics analysis demonstrates that the maximal efficacy is achieved at medium doses with drug exposure near saturating GITR occupancy during the dosing cycle. Finally, we propose an alternative dose-finding strategy that does not rely on the traditional maximal tolerated dose (MTD)-based paradigm but instead on utilizing the RO-function relations as biomarker to guide the clinical translation of GITR and similar co-stimulatory agonists.


Subject(s)
Glucocorticoids , Receptors, IgG , Cell Line, Tumor , Glucocorticoid-Induced TNFR-Related Protein/agonists , Ligands , Receptors, Tumor Necrosis Factor/agonists , Retrospective Studies , Tumor Necrosis Factors
6.
Cancer Lett ; 536: 215663, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35381307

ABSTRACT

Multiple myeloma (MM) is a hematological malignancy that results from the malignant proliferation of plasma cells in the bone marrow. B cell maturation antigen (BCMA) is highly selectively expressed in malignant plasma cells and is a novel therapeutic target for MM. Here, we developed a bispecific T cell engager, IBI379, that targets BCMA and CD3, and investigated its antitumor efficacy against MM. IBI379 showed strong binding affinity with both BCMA and CD3, which triggered T cell activation, proliferation, and cytokine release. An in vitro study demonstrated that IBI379 induced the lysis of MM cells expressing differing levels of BCMA on the cell surface. Administration of IBI379 in H929 or Daudi-BCMA cell xenograft mouse models significantly inhibited tumor growth without inducing body weight loss. The mechanism of action study revealed the accumulation of CD4+CD8+ T cells and granzyme B-positive T cells in tumors that were treated with IBI379. Moreover, administration of low dose of IBI379 in cynomolgus monkeys was well-tolerated and induced the depletion of BCMA+ B cells and a mild transient increase of cytokines. Collectively, these results demonstrate that IBI379 is a highly potent therapeutic strategy for depleting BCMA-positive B cells and is a promising approach for the treatment of MM.


Subject(s)
Antibodies, Bispecific , Multiple Myeloma , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , B-Cell Maturation Antigen/metabolism , CD3 Complex/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/therapeutic use , Humans , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Xenograft Model Antitumor Assays
7.
J Clin Invest ; 132(4)2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35108220

ABSTRACT

Many SARS-CoV-2 neutralizing antibodies (nAbs) lose potency against variants of concern. In this study, we developed 2 strategies to produce mutation-resistant antibodies. First, a yeast library expressing mutant receptor binding domains (RBDs) of the spike protein was utilized to screen for potent nAbs that are least susceptible to viral escape. Among the candidate antibodies, P5-22 displayed ultrahigh potency for virus neutralization as well as an outstanding mutation resistance profile. Additionally, P14-44 and P15-16 were recognized as mutation-resistant antibodies with broad betacoronavirus neutralization properties. P15-16 has only 1 binding hotspot, which is K378 in the RBD of SARS-CoV-2. The crystal structure of the P5-22, P14-44, and RBD ternary complex clarified the unique mechanisms that underlie the excellent mutation resistance profiles of these antibodies. Secondly, polymeric IgG enhanced antibody avidity by eliminating P5-22's only hotspot, residue F486 in the RBD, thereby potently blocking cell entry by mutant viruses. Structural and functional analyses of antibodies screened using both potency assays and the yeast RBD library revealed rare, ultrapotent, mutation-resistant nAbs against SARS-CoV-2.


Subject(s)
Antibodies, Viral/immunology , Broadly Neutralizing Antibodies/immunology , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/genetics , Antibody Affinity , B-Lymphocytes/immunology , Binding Sites/genetics , Binding Sites/immunology , Broadly Neutralizing Antibodies/blood , Broadly Neutralizing Antibodies/genetics , COVID-19/therapy , Cloning, Molecular , Disease Models, Animal , Humans , Immunization, Passive , Immunoglobulin G/immunology , In Vitro Techniques , Lung/virology , Mice , Mice, Inbred BALB C , Mutation , Neutralization Tests , Receptors, Virus/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , COVID-19 Serotherapy
8.
Stem Cell Res Ther ; 12(1): 525, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34620242

ABSTRACT

BACKGROUND: Globally, bone fractures are the most common musculoskeletal trauma, and approximately 8-10% of cases that fall into the categories of delayed or non-union healing. To date, there are no efficient pharmacological agents to accelerate the healing of bone fractures. Thus, it is necessary to find new strategies that accelerate bone healing and reduce the incidence of non-union or delayed fracture healing. Previous studies have revealed that the plasminogen activation system has been demonstrated to play an important role in bone metabolism. However, the function of SERPINB2 in the osteogenesis of hBMSCs remains unclear. Therefore, in this study, we investigated the effects and mechanism of SERPINB2 on osteogenic differentiation. METHODS: We investigated the osteogenesis effects of hBMSCs by both exogenous SerpinB2 protein and SERPINB2 gene silencing in vitro. Cell proliferation assay was used to assess the effect of exogenous SerpinB2 or SERPINB2 silencing on proliferation of hBMSCs. qPCR and Western blotting analysis detected the expression of target genes and proteins respectively. ALP staining was used to evaluated ALP activity and Alizarin Red staining (ARS) was used to evaluate mineral deposition. In vivo, a murie tibial fracture model was established, histological evaluation and radiographic analysis was used to confirm the therapeutic effects of SERPINB2 silencing in fracture healing. Statistical significance between two groups was determined by Student's t test, one-way ANOVA or Bonferroni's post-hoc test according to the distribution of the tested population. RESULTS: The addition of exogenous SerpinB2 protein inhibted osteoblast differentiation of hBMSCs in vitro, while SERPINB2 gene silencing significant promote osteoblast differentiation of hBMSCs in vitro. And silenced SERPINB2 gene also increased mineral deposits. Moreover, ß-catenin levels were up-regulated by SERPINB2 gene depletion. And the enhancement of osteogenic differentiation induced by SERPINB2 silencing was almost inhibited by specific Wnt/ß-catenin signaling pathway inhibitor. In a murine tibial fracture model, local injection of SERPINB2 siRNA improved bone fracture healing. CONCLUSIONS: Taken together, these findings indicate that SERPINB2 silencing promoted osteogenic differentiation of BMSCs via the Wnt/ß-catenin signaling pathway, and silenced SERPINB2 in vivo effectively promotes fracture healing, suggesting that SERPINB2 may be a novel target for bone fracture healing.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Mesenchymal Stem Cells , Osteogenesis , Wnt Signaling Pathway , Animals , Bone Marrow Cells/metabolism , Cell Differentiation , Cells, Cultured , Fractures, Bone/therapy , Gene Silencing , Humans , Mesenchymal Stem Cells/metabolism , Mice , Osteogenesis/genetics , beta Catenin/genetics , beta Catenin/metabolism
9.
Front Pharmacol ; 12: 607635, 2021.
Article in English | MEDLINE | ID: mdl-33935702

ABSTRACT

Glycyrrhizic acid (GA) is a major triterpene glycoside isolated from liquorice root that has been shown to inhibit osteoclastogenesis. However, there have been no reports regarding the effect of GA on osteogenic differentiation. Therefore, this study was performed to explore the effects and mechanism of action of GA on osteogenesis. A CCK-8 array was used to assess cell viability. The osteogenic capability was investigated by real-time quantitative PCR, western blotting and immunofluorescence analyses. ALP staining and ARS were used to evaluate ALP activity and mineralization, respectively. GA-GelMA hydrogels were designed to verify the therapeutic effects of GA in vivo by radiographic analysis and histological evaluation. Our results show that GA had no significant influence on the viability or proliferation of human bone marrow stromal cells (hBMSCs). GA promoted osteogenic differentiation and enhanced calcium deposition. Furthermore, ratio of active ß-catenin and total ß-catenin protein increased after treatment with GA. Wnt/catenin signaling inhibitor partially attenuated the effects of GA on osteogenic differentiation. In a mouse femoral fracture model, GA-GelMA hydrogels accelerated bone healing. Our results show that GA promotes the osteogenic differentiation of hBMSCs by modulating the Wnt/ß-catenin signaling pathway. GA-GelMA hydrogels promoted bone fracture healing. GA has potential as a cost-effective treatment of bone defects.

10.
Stem Cell Res Ther ; 12(1): 268, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33947456

ABSTRACT

BACKGROUND: Inflammatory microenvironment is significant to the differentiation and function of mesenchymal stem cells (MSCs). It evidentially influences the osteoblastogenesis of MSCs. IL-34, a newly discovered cytokine, playing a key role in metabolism. However, the research on its functional role in the osteogenesis of MSCs was rarely reported. Here, we described the regulatory effects of low-dose IL-34 on both osteoblastogenesis and osteoclastogenesis. METHODS: We performed the osteogenic effects of hBMSCs by exogenous and overexpressed IL-34 in vitro, so were the osteoclastogenesis effects of mBMMs by extracellular IL-34. CCK-8 was used to assess the effect of IL-34 on the viability of hBMSCs and mBMMs. ALP, ARS, and TRAP staining was used to evaluate ALP activity, mineral deposition, and osteoclastogenesis, respectively. qRT-PCR and Western blotting analysis were performed to detect the expression of target genes and proteins. ELISA was used to evaluate the concentrations of IL-34. In vivo, a rat tibial osteotomy model and an OVX model were established. Radiographic analysis and histological evaluation were performed to confirm the therapeutic effects of IL-34 in fracture healing and osteoporosis. Statistical differences were evaluated by two-tailed Student's t test, one-way ANOVA with Bonferroni's post hoc test, and two-way ANOVA with Bonferroni multiple comparisons post hoc test in the comparison of 2 groups, more than 2 groups, and different time points of treated groups, respectively. RESULTS: Promoted osteoblastogenesis of hBMSCs was observed after treated by exogenous or overexpressed IL-34 in vitro, confirmed by increased mineral deposits and ALP activity. Furthermore, exogenous or overexpressed IL-34 enhanced the expression of p-AKT and p-ERK. The specific AKT and ERK signaling pathway inhibitors suppressed the enhancement of osteoblastogenesis induced by IL-34. In a rat tibial osteotomy model, imaging and histological analyses testified the local injection of exogenous IL-34 improved bone healing. However, the additional IL-34 has no influence on both osteoclastogenesis of mBMMs in vitro and osteoporosis of OVX model of rat in vivo. CONCLUSIONS: Collectively, our study demonstrate that low-dose IL-34 regulates osteogenesis of hBMSCs partly via the PIK/AKT and ERK signaling pathway and enhances fracture healing, with neither promoting nor preventing osteoclastogenesis in vitro and osteoporosis in vivo.


Subject(s)
Osteogenesis , Proto-Oncogene Proteins c-akt , Animals , Cell Differentiation , Cells, Cultured , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction
11.
Inflammation ; 44(2): 434-449, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33215255

ABSTRACT

Continuous stimulation of inflammation is harmful to tissues of an organism. Inflammatory mediators not only have an effect on metabolic and inflammatory bone diseases but also have an adverse effect on certain genetic and periodontal diseases associated with bone destruction. Inflammatory factors promote vascular calcification in various diseases. Vascular calcification is a pathological process similar to bone development, and vascular diseases play an important role in the loss of bone homeostasis. The NLRP3 inflammasome is an essential component of the natural immune system. It can recognize pathogen-related molecular patterns or host-derived dangerous signaling molecules, recruit, and activate the pro-inflammatory protease caspase-1. Activated caspase-1 cleaves the precursors of IL-1ß and IL-18 to produce corresponding mature cytokines or recognizes and cleaves GSDMD to mediate cell pyroptosis. In this review, we discuss the role of NLRP3 inflammasome in bone diseases and vascular calcification caused by sterile or non-sterile inflammation and explore potential treatments to prevent bone loss.


Subject(s)
Bone Diseases/immunology , Inflammasomes/immunology , Inflammation/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Vascular Calcification/immunology , Biomarkers/metabolism , Bone Diseases/metabolism , Humans , Inflammasomes/metabolism , Inflammation/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Vascular Calcification/metabolism
12.
Int Immunopharmacol ; 89(Pt B): 107008, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33069927

ABSTRACT

Psoriasis is a highly prevalent inflammatory skin disease. Plaque psoriasis is the most common type of psoriasis, and the interleukin (IL)-23/IL-17 axis plays a key role in disease progression. In this article, we describe IBI112, a highly potent anti-IL-23 monoclonal antibody under clinical development, which efficiently neutralizes IL23p19, a subunit of IL-23, to abrogate IL-23 binding to its receptor and block downstream signal transducer and activator of transcription 3 (STAT3) phosphorylation. Specifically, IBI112 blocked IL-23 induced downstream IL-17 production from splenocytes. In addition, IBI112 administration reduced skin thickness in a psoriasis-like epidermal hyperplasia mouse model challenged by continuous hIL-23 injection. IBI112 showed synergism with an anti-IL-1R antibody in controlling disease progression in an imiquimod (IMQ) -induced psoriasis model. Moreover, with mutations in Fc fragment of IBI112, extended half-life was observed when compared to the wild-type IgG1 version in both human-FcRn-knock-in mice and cynomolgus monkeys. IBI112 was well tolerated after high dose administration in cynomolgus monkeys. In summary, we have developed an extended half-life, anti-IL-23p19 monoclonal antibody, IBI112, which efficiently neutralized IL-23, blocked IL-23-induced IL-17 production, and alleviated disease symptoms in two mouse models of psoriasis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Interleukin-23 Subunit p19/antagonists & inhibitors , Leukocytes, Mononuclear/drug effects , Psoriasis/drug therapy , Skin/drug effects , Animals , Anti-Inflammatory Agents/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacology , Cells, Cultured , Disease Models, Animal , Gene Knock-In Techniques , Half-Life , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Imiquimod , Interleukin-17/metabolism , Interleukin-23 , Interleukin-23 Subunit p19/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Macaca fascicularis , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Phosphorylation , Psoriasis/chemically induced , Psoriasis/immunology , Psoriasis/metabolism , Receptors, Fc/genetics , Receptors, Fc/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Skin/immunology , Skin/metabolism
13.
Mol Cancer Ther ; 19(12): 2564-2574, 2020 12.
Article in English | MEDLINE | ID: mdl-32999045

ABSTRACT

Immunotherapy using OX40 agonist antibodies shows great preclinical efficacy in mouse tumor models. But in a clinical setting, OX40 agonist antibody alone or in combination with checkpoint blockade exhibits only modest efficacy due to lack of sufficient activation. We hypothesized that the limited antitumor activity in patients may due to insufficient clustering of OX40 antibody in the tumor. To test this hypothesis, we generated a tetravalent programmed death ligand-1 (PD-L1)/OX40 BsAb by fusing two PD-L1 VHH fragments to the C-terminus of a nonblocking agonistic anti-OX40 antibody. The resulting BsAb had intact function of each parental antibody, including efficiently blocking PD1/PD-L1 interaction and inducing OX40 activation. In addition, this BsAb showed significantly enhanced potency in activation of OX40-expressing T cells when PD-L1-expressing tumor cells or dendrite cells were present, through PD-L1-mediated cross-linking of OX40. Moreover, the BsAb exhibited superior antitumor activities over the parental monospecific antibodies alone or in combination in multiple in vivo tumor models. These results demonstrated a great potential for further clinical development of the potent immunostimulatory PD-L1/OX40 bispecific antibody.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Receptors, OX40/agonists , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Immunomodulation/drug effects , Mice , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
14.
Int Immunopharmacol ; 88: 106960, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32919219

ABSTRACT

BACKGROUND: The treatment of bone defects has always been a problem for clinicians. In recent years, research on human bone mesenchymal stem cells (hBMSCs) has found that promoting their osteogenic differentiation could be a useful therapeutic strategy for bone healing. Previous studies have been reported that Withania somnifera Dunal inhibits osteoclastogenesis by inhibiting the NF-κB signaling pathway. Withanolide B is an active component of W. somnifera Dunal, but its role in osteogenic differentiation of hBMSCs remains unknown. Here, we performed a preliminary study on the role of Withanolide B in promoting osteogenic differentiation and its possible mechanism. METHODS: We investigated the effect of Withanolide B on osteogenic differentiation of hBMSCs in vitro and in vivo. The effect of Withanolide B on the activity of hBMSCs was verified by CCK-8 assay and quantitative Real-time polymerase chain reaction (qPCR) and Western blotting analysis were used to verify the effect of Withanolide B on osteogenic differentiation-specific genes and proteins. The effect of Withanolide B on ALP activity and mineral deposition was verified by ALP and ARS staining. We then used a rat tibial osteotomy model to observe the effect of Withanolide B on bone healing. RESULTS: Withanolide B is noncytotoxic to hBMSCs and can effectively promote their osteogenic differentiation. Moreover, we found that Withanolide B can regulate the osteogenic differentiation of hBMSCs through the ERK1/2 and Wnt/ß-catenin signaling pathways. When inhibitors of the ERK1/2 and Wnt/ß-catenin signaling pathways were used, the enhancement of osteogenic differentiation induced by Withanolide B was attenuated. Withanolide B also effectively promoted bone healing in the rat tibial osteotomy model. CONCLUSIONS: Our results suggest that Withanolide B can promote the osteogenic differentiation of hBMSCs through the ERK1/2 and Wnt/ß-catenin signaling pathways and can effectively promote bone defect healing.


Subject(s)
Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Withanolides/pharmacology , Animals , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Cells, Cultured , Humans , MAP Kinase Signaling System/drug effects , Male , Rats, Sprague-Dawley , Tibia/drug effects , Tibia/injuries , Wnt Signaling Pathway/drug effects
15.
J Cell Mol Med ; 24(18): 10444-10457, 2020 09.
Article in English | MEDLINE | ID: mdl-32681612

ABSTRACT

Over-activated osteoclastogenesis, which is initiated by inflammation, has been implicated in osteoporosis. Corilagin, a natural compound extracted from various medicinal herbaceous plants, such as Cinnamomum cassia, has antioxidant and anti-inflammatory activities. We found that Corilagin suppressed osteoclast differentiation in a dose-dependent manner, significantly decreased osteoclast-related gene expression and impaired bone resorption by osteoclasts. Moreover, phosphorylation of members of the nuclear factor-kappaB (NF-κB) and PI3K/AKT signalling pathways was reduced by Corilagin. In a murine model of osteoporosis, Corilagin inhibited osteoclast functions in vivo and restored oestrogen deficiency-induced bone loss. In conclusion, our findings suggested that Corilagin inhibited osteoclastogenesis by down-regulating the NF-κB and PI3K/AKT signalling pathways, thus showing its potential possibility for the treatment of osteoporosis.


Subject(s)
Bone Resorption/pathology , Estrogens/deficiency , Glucosides/pharmacology , Hydrolyzable Tannins/pharmacology , NF-kappa B/metabolism , Osteogenesis/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RANK Ligand/pharmacology , Actins/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Down-Regulation/drug effects , Glucosides/chemistry , Hydrolyzable Tannins/chemistry , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Osteoprotegerin/metabolism , Ovariectomy , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
16.
Int Immunopharmacol ; 84: 106550, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32388216

ABSTRACT

The metabolicosteopathy known as postmenopausal osteoporosisiscaused by disruption of the balance between bone resorption and osteogenesis, processes that are mediated by osteoclasts and osteoblasts, respectively. The current therapeutic approaches to treating osteoporosis have several limitations. In this study, we demonstrated that the natural chemical compound isoalantolactone (IAL) could inhibit osteoclastogenesis, without affecting osteogenesis. This is the first study reporting a role of IAL in suppressing the receptor activator of nuclear factor-kappa B ligand (RANKL)-induced osteoclast formation in a dose-dependent manner, and downregulating the expression of osteoclast-related marker genes. Furthermore, IAL abrogated the phosphorylation of c-Jun N-terminal kinase (JNK)/p38, NF-κB, and phosphatidylinositol 3-kinase (PI3K)-AKT, and also diminished the expression of osteoclastogenesis-related proteins. In conclusion, our results indicated that IAL has promise for the treatment of osteoporosis and other metabolicbone diseases.


Subject(s)
Bone Resorption/drug therapy , Osteoclasts/drug effects , Osteogenesis/drug effects , Sesquiterpenes/therapeutic use , Actins/metabolism , Animals , Cathepsin K/genetics , Cathepsin K/metabolism , Cells, Cultured , Female , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Ovariectomy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-fos/metabolism , RANK Ligand , Sesquiterpenes/pharmacology , Signal Transduction/drug effects
17.
J Shoulder Elbow Surg ; 29(5): 996-1001, 2020 May.
Article in English | MEDLINE | ID: mdl-32305108

ABSTRACT

BACKGROUND: Heterotopic ossification (HO) is a common complication after surgery for elbow trauma. Uric acid is the end product of purine metabolism and has several physiological and pathogenic roles. However, the relationship between HO and uric acid has not been explored. This retrospective study aimed to assess the relationship between HO and serum uric acid (SUA). MATERIAL AND METHODS: We retrospectively reviewed data from 155 patients undergoing elbow trauma surgery in our hospital between January 2013 and December 2018. One hundred patients were included according to the inclusion criteria. They were divided into 2 groups according to the presence or absence of HO, and the SUA level was compared between groups using the independent samples t test. The optimal prognostic cutoff value was obtained using the maximum value of the Youden index. RESULTS: The SUA level was significantly higher in the HO group than in the non-HO group (362.0 ± 87.4 µmol/L vs. 318.3 ± 87.0 µmol/L; P < .05). Using the maximum value of Youden index, 317.5 µmol/L was determined to be the optimal SUA cutoff value for the prediction of HO, with a sensitivity of 68.75% (95% confidence interval [CI], 54.67%-80.05%) and specificity of 55.77% (95% CI, 42.34%-68.40%). CONCLUSIONS: Our study was the first to find that the high SUA level is a risk factor for HO of the elbow joint after trauma. Moreover, 317.5 µmol/L is the SUA threshold predicting the occurrence and development of HO of the elbow, with high sensitivity and specificity.


Subject(s)
Elbow Joint/surgery , Elbow/surgery , Ossification, Heterotopic/blood , Ossification, Heterotopic/etiology , Uric Acid/blood , Adult , Aged , Female , Humans , Male , Middle Aged , Postoperative Complications/blood , Postoperative Complications/etiology , Prognosis , Range of Motion, Articular , Retrospective Studies , Risk Factors , Sensitivity and Specificity , Time Factors , Young Adult , Elbow Injuries
18.
Cancer Immunol Immunother ; 69(6): 939-950, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32078015

ABSTRACT

With the great success of anti-CTLA-4 and anti-PD-1 therapeutics in cancer immunotherapy, tumor necrosis factor receptor superfamily members have been recognized as ideal targets to provide co-stimulatory signals in combination with immune checkpoint blocking antibodies. Among these is OX40 (CD134), a co-stimulatory molecule expressed by activated immune cells. Recently, several anti-OX40 agonistic monoclonal antibodies, pogalizumab as the most advanced, have entered early phase clinical trials. Using a yeast platform and multiple screening methods, we identified a fully human anti-OX40 antibody (IBI101) with distinct modes of action. Unlike pogalizumab, IBI101 partially blocks the binding of OX40 to its ligand OX40L and exhibits both FcγR-dependent and independent agonistic activities in NF-κB luciferase reporter assays. IBI101 also promotes T cell activation and proliferation in vitro. These unique properties partially explain the more potent anti-tumor activity of IBI101 than that of pogalizumab in humanized NOG mice bearing LoVo tumors. In addition, IBI101 shows efficacious anti-tumor activity in mice when administrated alone or in combination with anti-PD-1 antibodies. In human OX40 knock-in mice bearing MC38 colon carcinoma, IBI101 treatment induces tumor antigen-specific CD8+ T-cell responses, decreases immunosuppressive regulatory T cells in tumor, and enhances the immune response to PD-1 inhibition. Preclinical studies of IBI101 in non-human primates demonstrate typical pharmacokinetic characteristics of an IgG antibody and no drug-related toxicity. Collectively, IBI101 has desirable preclinical attributes which support its clinical development for cancer treatment.


Subject(s)
Immunotherapy/methods , Receptors, OX40/immunology , Animals , Cell Line , Disease Models, Animal , Humans , Mice
19.
Antib Ther ; 3(4): 227-236, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33928230

ABSTRACT

BACKGROUND: Strategies to reinvigorate exhausted T cells have achieved great efficacy in certain subpopulations of tumor patients. Blocking the antibodies that target programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 induces durable responses in Hodgkin's lymphoma, melanoma, renal and lung cancers. T cell immunoglobulin mucin-3 (TIM-3) is another well-defined inhibitory receptor that is expressed in terminally differentiated Th1/Tc1 cells, which produces interferon gamma and cytotoxic molecules. It is also significantly expressed on forkhead box P3+ regulatory T cells and innate immune cells such as dendritic cells and macrophages. METHODS: By immunizing BALB/c mice with recombinant TIM-3 and screening of 20 000 hybridoma clones, we selected a monoclonal TIM-3-blocking antibody (IBI104), which shows great efficacy in vitro and in vivo. RESULTS: IBI104 blocks phosphatidylserine interaction with TIM-3 but does not interfere with the interaction of TIM-3 with galectin-9 in ELISA assays. However, in vitro administration of IBI104 induces the potent internalization of TIM-3 in activated T cells to the extent that it will shut down the entire TIM-3 mediated signaling regardless of the ligands. IBI104 shows potent anti-tumor efficacy when combined with anti-PD1 in vivo. CONCLUSIONS: Our results suggest that IBI104 is a promising blocking antibody for TIM-3-mediated suppressive signaling and can serve as effective cancer immunotherapy, especially in combination with anti-PD1.

20.
Biomed Res Int ; 2019: 3456719, 2019.
Article in English | MEDLINE | ID: mdl-31534957

ABSTRACT

Dihydroartemisinin (DHA), which is considered to be one of the active compounds within Artemisia annua, has extensively been used in recent years as the most effective drug against malaria, having many biological functions including anticancer, antifungal, and immunomodulatory activities. However, DHA plays a role in the regulation of the proliferation and human mesenchymal stem cells (hMSCs) osteogenic differentiation that remains unknown. We explored DHA's effect on hMSCs' proliferation as well as the osteogenic differentiation, together with its underlying mechanisms of action. We showed that DHA enhanced osteogenic differentiation but had no significant effect on hMSCs' proliferation. It probably exerted its functions through the signaling pathways of ERK1/2 as well as Wnt/ß. Because DHA has low toxicity and costs, it might be regarded as an important drug for fracture treatment and tissue engineering.


Subject(s)
Artemisinins/pharmacology , Cell Differentiation/drug effects , MAP Kinase Signaling System/drug effects , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Wnt Signaling Pathway/drug effects , Cell Proliferation/drug effects , Humans , Mesenchymal Stem Cells/cytology , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...