Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Mol Med (Berl) ; 102(5): 693-707, 2024 05.
Article in English | MEDLINE | ID: mdl-38492027

ABSTRACT

Physical therapy is extensively employed in clinical settings. Nevertheless, the absence of suitable animal models has resulted in an incomplete understanding of the in vivo mechanisms and cellular distribution that respond to physical stimuli. The objective of this research was to create a mouse model capable of indicating the cells affected by physical stimuli. In this study, we successfully established a mouse line based on the heat shock protein 70 (Hsp70) promoter, wherein the expression of CreERT2 can be induced by physical stimuli. Following stimulation of the mouse tail, ear, or cultured calvarias with heat shock (generated by heating, ultrasound, or laser), a distinct Cre-mediated excision was observed in cells stimulated by these physical factors with minimal occurrence of leaky reporter expression. The application of heat shock to Hsp70-CreERT2; FGFR2-P253R double transgenic mice or Hsp70-CreERT2 mice infected with AAV-BMP4 at calvarias induced the activation of Cre-dependent mutant FGFR2-P253R or BMP4 respectively, thereby facilitating the premature closure of cranial sutures or the repair of calvarial defects. This novel mouse line holds significant potential for investigating the underlying mechanisms of physical therapy, tissue repair and regeneration, lineage tracing, and targeted modulation of gene expression of cells in local tissue stimulated by physical factor at the interested time points. KEY MESSAGES: In the study, an Hsp70-CreERT2 transgenic mouse was generated for heat shock-induced gene modulation. Heat shock, ultrasound, and laser stimulation effectively activated Cre expression in Hsp70-CreERT2; reporter mice, which leads to deletion of floxed DNA sequence in the tail, ear, and cultured calvaria tissues of mice. Local laser stimuli on cultured calvarias effectively induce Fgfr2-P253R expression in Hsp70-mTmG-Fgfr2-P253R mice and result in accelerated premature closure of cranial suture. Heat shock activated AAV9-FLEX-BMP4 expression and subsequently promoted the repair of calvarial defect of Hsp70-CreERT2; Rosa26-mTmG mice.


Subject(s)
Bone Morphogenetic Protein 4 , HSP70 Heat-Shock Proteins , Mice, Transgenic , Promoter Regions, Genetic , Animals , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Mice , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Heat-Shock Response/genetics , Skull/metabolism , Gene Expression Regulation , Integrases/metabolism , Integrases/genetics
2.
Adv Sci (Weinh) ; 11(7): e2306143, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38083984

ABSTRACT

Macrophages are heterogenic phagocytic cells that play distinct roles in physiological and pathological processes. Targeting different types of macrophages has shown potent therapeutic effects in many diseases. Although many approaches are developed to target anti-inflammatory macrophages, there are few researches on targeting pro-inflammatory macrophages, which is partially attributed to their non-s pecificity phagocytosis of extracellular substances. In this study, a novel recombinant protein is constructed that can be anchored on an exosome membrane with the purpose of targeting pro-inflammatory macrophages via antigen recognition, which is named AnCar-ExoLaIMTS . The data indicate that the phagocytosis efficiencies of pro-inflammatory macrophages for different AnCar-ExoLaIMTS show obvious differences. The AnCar-ExoLaIMTS3 has the best targeting ability for pro-inflammatory macrophages in vitro and in vivo. Mechanically, AnCar-ExoLaIMTS3 can specifically recognize the leucine-rich repeat domain of the TLR4 receptor, and then enter into pro-inflammatory macrophages via the TLR4-mediated receptor endocytosis pathway. Moreover, AnCar-ExoLaIMTS3 can efficiently deliver therapeutic cargo to pro-inflammatory macrophages and inhibit the synovial inflammatory response via downregulation of HIF-1α level, thus ameliorating the severity of arthritis in vivo. Collectively, the work established a novel gene/drug delivery system that can specifically target pro-inflammatory macrophages, which may be beneficial for the treatments of arthritis and other inflammatory diseases.


Subject(s)
Arthritis , Macrophages , Humans , Macrophages/metabolism , Arthritis/drug therapy , Phagocytosis , Anti-Inflammatory Agents/therapeutic use , Cell Communication
3.
Stem Cell Res Ther ; 13(1): 227, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35659742

ABSTRACT

BACKGROUND: Intervertebral disc degeneration (IVDD) can cause low back pain, a major public health concern. IVDD is characterized with loss of cells especially those in nucleus pulposus (NP), due to the limited proliferative potential and regenerative ability. Few studies, however, have been carried out to investigate the in vivo proliferation events of NP cells and the cellular contribution of a specific subpopulation of NP during postnatal growth or regeneration. METHODS: We generated FGFR3-3*Flag-IRES-GFP mice and crossed FGFR3-CreERT2 mice with Rosa26-mTmG, Rosa26-DTA and Rosa26-Confetti mice, respectively, to perform inducible genetic tracing studies. RESULTS: Expression of FGFR3 was found in the outer region of NP with co-localized expressions of proliferating markers. By fate mapping studies, FGFR3-positive (FGFR3+) NP cells were found proliferate from outer region to inner region of NP during postnatal growth. Clonal lineage tracing by Confetti mice and ablation of FGFR3·+ NP cells by DTA mice further revealed that the expansion of the FGFR3+ cells was required for the morphogenesis and homeostasis of postnatal NP. Moreover, in degeneration and regeneration model of mouse intervertebral disc, FGFR3+ NP cells underwent extensive expansion during the recovery stage. CONCLUSION: Our present work demonstrates that FGFR3+ NP cells are novel subpopulation of postnatal NP with long-existing proliferative capacity shaping the adult NP structure and participating in the homeostasis maintenance and intrinsic repair of NP. These findings may facilitate the development of new therapeutic approaches for IVD regeneration.


Subject(s)
Intervertebral Disc Degeneration , Intervertebral Disc , Low Back Pain , Nucleus Pulposus , Animals , Cells, Cultured , Intervertebral Disc Degeneration/therapy , Mice , Nucleus Pulposus/metabolism
4.
Int J Biol Sci ; 17(15): 4140-4153, 2021.
Article in English | MEDLINE | ID: mdl-34803488

ABSTRACT

Systemic application of glucocorticoids is an essential anti-inflammatory and immune-modulating therapy for severe inflammatory or autoimmunity conditions. However, its long-term effects on articular cartilage of patients' health need to be further investigated. In this study, we studied the effects of dexamethasone (Dex) on the homeostasis of articular cartilage and the progress of destabilization of medial meniscus (DMM)-induced osteoarthritis (OA) in adult mice. Long-term administration of Dex aggravates the proteoglycan loss of articular cartilage and drastically accelerates cartilage degeneration under surgically induced OA conditions. In addition, Dex increases calcium content in calcified cartilage layer of mice and the samples from OA patients with a history of long-term Dex treatment. Moreover, long term usage of Dex results in decrease subchondral bone mass and bone density. Further studies showed that Dex leads to calcification of extracellular matrix of chondrocytes partially through activation of AKT, as well as promotes apoptosis of chondrocytes in calcified cartilage layer. Besides, Dex weakens the stress-response autophagy with the passage of time. Taken together, our data indicate that long-term application of Dex may predispose patients to OA and or even accelerate the OA disease progression development of OA patients.


Subject(s)
Apoptosis/drug effects , Chondrocytes/drug effects , Chondrocytes/physiology , Dexamethasone/adverse effects , Extracellular Matrix/drug effects , Osteoarthritis/etiology , Animals , Calcinosis , Dexamethasone/administration & dosage , Drug Administration Schedule , Glucocorticoids/administration & dosage , Glucocorticoids/adverse effects , Homeostasis , Male , Mice , Mice, Inbred C57BL , Osteoarthritis/pathology
5.
ACS Nano ; 15(10): 15874-15891, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34586802

ABSTRACT

The clinical application of small interfering RNA (siRNA) drugs provides promising opportunities to develop treatment strategies for autoimmune inflammatory diseases. In this study, siRNAs targeting the endoplasmic reticulum to nucleus signaling 1 (ERN1) gene (siERN1) were screened. Two cationic polymers, polyethylenimine (PEI) and poly(ß-amino amine) (PBAA), which can improve the efficiency of the siRNA transfection, were used as siERN1 delivery carriers. They were implemented to construct a nanodrug delivery system with macrophage-targeting ability and dual responsiveness for the treatment of autoimmune inflammatory diseases. In terms of the mechanism, siERN1 can regulate the intracellular calcium ion concentration by interfering with the function of inositol 1,4,5-trisphosphate receptor 1/3 (IP3R1/3) and thus inducing M2 polarization of macrophages. Furthermore, siERN1-nanoprodrug [FA (folic acid)-PEG-R(RKKRRQRRR)-NPs(ss-PBAA-PEI)@siERN1] acts as a conductor of macrophage polarization by controlling the calcium ion concentration and is an inhibitor of MyD88-dependent Toll-like receptor signaling. The results revealed that the FA-PEG-R-NPs@siERN1 has universal biocompatibility, long-term drug release responsiveness, superior targeting properties, and therapeutic effects in mouse collagen-induced arthritis and inflammatory bowel disease models. In conclusion, this study reveals a potential strategy to treat autoimmune inflammatory disorders.


Subject(s)
Polyethyleneimine , Toll-Like Receptors , Animals , Macrophages , Mice , RNA, Small Interfering , Transfection
6.
Nat Commun ; 12(1): 4391, 2021 07 19.
Article in English | MEDLINE | ID: mdl-34282140

ABSTRACT

Acquired heterotopic ossification (HO) is the extraskeletal bone formation after trauma. Various mesenchymal progenitors are reported to participate in ectopic bone formation. Here we induce acquired HO in mice by Achilles tenotomy and observe that conditional knockout (cKO) of fibroblast growth factor receptor 3 (FGFR3) in Col2+ cells promote acquired HO development. Lineage tracing studies reveal that Col2+ cells adopt fate of lymphatic endothelial cells (LECs) instead of chondrocytes or osteoblasts during HO development. FGFR3 cKO in Prox1+ LECs causes even more aggravated HO formation. We further demonstrate that FGFR3 deficiency in LECs leads to decreased local lymphatic formation in a BMPR1a-pSmad1/5-dependent manner, which exacerbates inflammatory levels in the repaired tendon. Local administration of FGF9 in Matrigel inhibits heterotopic bone formation, which is dependent on FGFR3 expression in LECs. Here we uncover Col2+ lineage cells as an origin of lymphatic endothelium, which regulates local inflammatory microenvironment after trauma and thus influences HO development via FGFR3-BMPR1a pathway. Activation of FGFR3 in LECs may be a therapeutic strategy to inhibit acquired HO formation via increasing local lymphangiogenesis.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Lymphatic Vessels/metabolism , Ossification, Heterotopic/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Achilles Tendon , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Endothelium, Lymphatic/metabolism , Gene Knockdown Techniques , Lymphangiogenesis , Male , Mesenchymal Stem Cells , Mice , Tenotomy
7.
J Cell Physiol ; 236(7): 5278-5292, 2021 07.
Article in English | MEDLINE | ID: mdl-33452687

ABSTRACT

Osteoarthritis (OA) is the most common joint disease. The surface of joint cartilage is a defensive and first affected structure of articular cartilage (AC) during the pathogenesis of OA. Alk5 signaling is critical for maintaining AC homeostasis, however, the role and underlying mechanism for the involvement of Alk5 signaling in the phenotypes of articular cartilage stem cells (ACSCs) at the surface of AC is still unclear. The role of Alk5 in OA development was explored using an ACSCs-specific Alk5-deficient (cKO) mouse model. Alterations in cartilage structure were evaluated histologically. Senescence was detected by SA-ß-gal, while reactive oxygen species (ROS), MitoTracker, and LysoTracker staining were used to detect changes related to senescence. In addition, mice were injected intra-articularly with ganciclovir to limit the detrimental roles of senescent cells (SnCs). Alk5 cKO mice showed a decreased number of the slow-cell cycle cells and less lubricant secretion at the surface accompanied with drastically accelerated cartilage degeneration under ageing and surgically induced OA conditions. Further studies showed that Alk5 deficient ACSCs exhibited senescence-like manifestations including decreased proliferation and differentiation, more SA-ß-gal-positive cells and ROS production, as well as significantly swollen mitochondria and lysosome breakdown. We further found that local limitation of the detrimental roles of SnCs can attenuate the development of posttraumatic OA. Taken together, our findings suggest that Alk5 signaling acts as an important regulator of the SnCs in the superficial layer during AC maintenance and OA initiation.


Subject(s)
Cartilage, Articular/metabolism , Cellular Senescence/physiology , Osteoarthritis/metabolism , Receptor, Transforming Growth Factor-beta Type I/metabolism , Stem Cells/metabolism , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Cartilage, Articular/pathology , Mice , Mice, Knockout , Osteoarthritis/pathology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism
8.
Signal Transduct Target Ther ; 5(1): 181, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32879300

ABSTRACT

Growing evidences suggest that the fibroblast growth factor/FGF receptor (FGF/FGFR) signaling has crucial roles in a multitude of processes during embryonic development and adult homeostasis by regulating cellular lineage commitment, differentiation, proliferation, and apoptosis of various types of cells. In this review, we provide a comprehensive overview of the current understanding of FGF signaling and its roles in organ development, injury repair, and the pathophysiology of spectrum of diseases, which is a consequence of FGF signaling dysregulation, including cancers and chronic kidney disease (CKD). In this context, the agonists and antagonists for FGF-FGFRs might have therapeutic benefits in multiple systems.


Subject(s)
Embryonic Development/genetics , Fibroblast Growth Factors/genetics , Homeostasis/genetics , Receptors, Fibroblast Growth Factor/genetics , Apoptosis/genetics , Cell Differentiation/genetics , Cell Proliferation , Humans , Neoplasms/genetics , Signal Transduction/genetics
9.
Theranostics ; 10(16): 7111-7130, 2020.
Article in English | MEDLINE | ID: mdl-32641982

ABSTRACT

CATSHL syndrome, characterized by camptodactyly, tall stature and hearing loss, is caused by loss-of-function mutations of fibroblast growth factor receptors 3 (FGFR3) gene. Most manifestations of patients with CATSHL syndrome start to develop in the embryonic stage, such as skeletal overgrowth, craniofacial abnormalities, however, the pathogenesis of these phenotypes especially the early maldevelopment remains incompletely understood. Furthermore, there are no effective therapeutic targets for this skeleton dysplasia. Methods: We generated fgfr3 knockout zebrafish by CRISPR/Cas9 technology to study the developmental mechanisms and therapeutic targets of CATSHL syndrome. Several zebrafish transgenic lines labeling osteoblasts and chondrocytes, and live Alizarin red staining were used to analyze the dynamical skeleton development in fgfr3 mutants. Western blotting, whole mount in situ hybridization, Edu labeling based cell proliferation assay and Wnt/ß-catenin signaling antagonist were used to explore the potential mechanisms and therapeutic targets. Results: We found that fgfr3 mutant zebrafish, staring from early development stage, showed craniofacial bone malformation with microcephaly and delayed closure of cranial sutures, chondroma-like lesion and abnormal development of auditory sensory organs, partially resembling the clinical manifestations of patients with CATSHL syndrome. Further studies showed that fgfr3 regulates the patterning and shaping of pharyngeal arches and the timely ossification of craniofacial skeleton. The abnormal development of pharyngeal arch cartilage is related to the augmented hypertrophy and disordered arrangement of chondrocytes, while decreased proliferation, differentiation and mineralization of osteoblasts may be involved in the delayed maturation of skull bones. Furthermore, we revealed that deficiency of fgfr3 leads to enhanced IHH signaling and up-regulated canonical Wnt/ß-catenin signaling, and pharmacological inhibition of Wnt/ß-catenin could partially alleviate the phenotypes of fgfr3 mutants. Conclusions: Our study further reveals some novel phenotypes and underlying developmental mechanism of CATSHL syndrome, which deepens our understanding of the pathogenesis of CATSHL and the role of fgfr3 in skeleton development. Our findings provide evidence that modulation of Wnt/ß-catenin activity could be a potential therapy for CATSHL syndrome and related skeleton diseases.


Subject(s)
Bone Diseases, Developmental/genetics , Chondrocytes/pathology , Chondrogenesis/genetics , Hand Deformities, Congenital/genetics , Hearing Loss/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Skull/embryology , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Bone Diseases, Developmental/pathology , CRISPR-Cas Systems/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Disease Models, Animal , Embryo, Nonmammalian , Gene Knockout Techniques , Hand Deformities, Congenital/pathology , Hearing Loss/pathology , Hedgehog Proteins/metabolism , Humans , Mutation , Wnt Signaling Pathway/genetics , Zebrafish
10.
FEBS Open Bio ; 10(3): 434-443, 2020 03.
Article in English | MEDLINE | ID: mdl-31975545

ABSTRACT

Low-intensity pulsed ultrasound (LIPUS), a noninvasive physical therapy, was recently demonstrated to be an effective treatment for osteoarthritis (OA). Vascular endothelium growth factor A (VEGFA) has been found to be upregulated in the articular cartilage, synovium and subchondral bone of OA patients, leading to cartilage degeneration, synovitis and osteophyte formation. However, the functions and mechanisms of LIPUS in regulating chondrocyte-derived VEGFA expression are still unclear. In this study, we investigated whether LIPUS attenuated OA progression by (a) decreasing the percentage of VEGFA-positive cells in mouse articular cartilage destabilised through medial meniscus surgery and (b) relieving interleukin-1ß-induced VEGFA expression in mouse primary chondrocytes. However, this function was negated by a p38 mitogen-activated protein kinase (p38 MAPK) inhibitor. In addition, we found that LIPUS ameliorated VEGFA-mediated disorders in cartilage extracellular matrix metabolism and chondrocyte hypertrophy during OA development. In conclusion, our data indicate a novel effect of LIPUS in regulating the expression of osteoarthritic chondrocyte-derived VEGFA through the suppression of p38 MAPK activity.


Subject(s)
Cartilage/metabolism , Chondrocytes/metabolism , Ultrasonic Therapy/methods , Vascular Endothelial Growth Factor A/metabolism , Animals , Cartilage/physiology , Cartilage Diseases/metabolism , Cartilage, Articular/metabolism , Extracellular Matrix/metabolism , Male , Mice , Mice, Inbred C57BL , Osteoarthritis/metabolism , Osteoarthritis/physiopathology , Osteophyte/metabolism , Protective Agents/metabolism , Protective Agents/pharmacology , Synovitis/metabolism , Ultrasonic Waves , Vascular Endothelial Growth Factor A/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/pharmacology
11.
Ann Rheum Dis ; 79(1): 112-122, 2020 01.
Article in English | MEDLINE | ID: mdl-31662319

ABSTRACT

OBJECTIVES: This study aims to investigate the role and mechanism of FGFR3 in macrophages and their biological effects on the pathology of arthritis. METHODS: Mice with conditional knockout of FGFR3 in myeloid cells (R3cKO) were generated. Gait behaviours of the mice were monitored at different ages. Spontaneous synovial joint destruction was evaluated by digital radiographic imaging and µCT analysis; changes of articular cartilage and synovitis were determined by histological analysis. The recruitment of macrophages in the synovium was examined by immunostaining and monocyte trafficking assay. RNA-seq analysis, Western blotting and chemotaxis experiment were performed on control and FGFR3-deficient macrophages. The peripheral blood from non-osteoarthritis (OA) donors and patients with OA were analysed. Mice were treated with neutralising antibody against CXCR7 to investigate the role of CXCR7 in arthritis. RESULTS: R3cKO mice but not control mice developed spontaneous cartilage destruction in multiple synovial joints at the age of 13 months. Moreover, the synovitis and macrophage accumulation were observed in the joints of 9-month-old R3cKO mice when the articular cartilage was not grossly destructed. FGFR3 deficiency in myeloid cells also aggravated joint destruction in DMM mouse model. Mechanically, FGFR3 deficiency promoted macrophage chemotaxis partly through activation of NF-κB/CXCR7 pathway. Inhibition of CXCR7 could significantly reverse FGFR3-deficiency-enhanced macrophage chemotaxis and the arthritic phenotype in R3cKO mice. CONCLUSIONS: Our study identifies the role of FGFR3 in synovial macrophage recruitment and synovitis, which provides a new insight into the pathological mechanisms of inflammation-related arthritis.


Subject(s)
Cartilage, Articular/pathology , Chemokine CXCL12/metabolism , Macrophages/metabolism , Osteoarthritis/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptors, CXCR/genetics , Synovitis/genetics , Animals , Chemotaxis/genetics , Gait , Gene Expression Regulation , Humans , Joints/metabolism , Joints/pathology , Mice , Mice, Knockout , Monocytes/metabolism , Myeloid Cells , NF-kappa B/metabolism , Osteoarthritis/metabolism , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptors, CXCR/metabolism , Synovial Membrane/metabolism , Synovial Membrane/pathology , Synovitis/pathology
12.
J Bone Miner Res ; 34(11): 2101-2116, 2019 11.
Article in English | MEDLINE | ID: mdl-31237961

ABSTRACT

Cartilage-hair hypoplasia (CHH) is an autosomal recessive metaphyseal chondrodysplasia characterized by bone dysplasia and many other highly variable features. The gene responsible for CHH is the RNA component of the mitochondrial RNA-processing endoribonuclease (RMRP) gene. Currently, the pathogenesis of osteochondrodysplasia and extraskeletal manifestations in CHH patients remains incompletely understood; in addition, there are no viable animal models for CHH. We generated an rmrp KO zebrafish model to study the developmental mechanisms of CHH. We found that rmrp is required for the patterning and shaping of pharyngeal arches. Rmrp mutation inhibits the intramembranous ossification of skull bones and promotes vertebrae ossification. The abnormalities of endochondral bone ossification are variable, depending on the degree of dysregulated chondrogenesis. Moreover, rmrp mutation inhibits cell proliferation and promotes apoptosis through dysregulating the expressions of cell-cycle- and apoptosis-related genes. We also demonstrate that rmrp mutation upregulates canonical Wnt/ß-catenin signaling; the pharmacological inhibition of Wnt/ß-catenin could partially alleviate the chondrodysplasia and increased vertebrae mineralization in rmrp mutants. Our study, by establishing a novel zebrafish model for CHH, partially reveals the underlying mechanism of CHH, hence deepening our understanding of the role of rmrp in skeleton development.


Subject(s)
Chondrogenesis/genetics , Hair/abnormalities , Hirschsprung Disease , Mutation , Osteochondrodysplasias/congenital , Osteogenesis/genetics , Primary Immunodeficiency Diseases , RNA, Long Noncoding , Wnt Signaling Pathway/genetics , Zebrafish/metabolism , Animals , Disease Models, Animal , Hair/metabolism , Hair/pathology , Hirschsprung Disease/genetics , Hirschsprung Disease/metabolism , Hirschsprung Disease/pathology , Humans , Osteochondrodysplasias/genetics , Osteochondrodysplasias/metabolism , Osteochondrodysplasias/pathology , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/metabolism , Primary Immunodeficiency Diseases/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Skull/metabolism , Skull/pathology , Spine/metabolism , Spine/pathology
13.
Int J Biol Sci ; 14(10): 1389-1398, 2018.
Article in English | MEDLINE | ID: mdl-30123084

ABSTRACT

It has been reported that overactivation of fibroblast growth factor receptor 1 (FGFR1) is an important characteristic found in most non-small cell lung cancer (NSCLC) samples. Here, we identified a FGFR1 inhibitory peptide R1-P2 and investigated its effects on the lung cancer cells growth and angiogenesis in vitro and in vivo. Our results demonstrate that R1-P2 bound to human FGFR1 protein, and efficiently blocked the binding of FGF2 to FGFR1 in A549 and NCI-H460 cells. Moreover, this peptide significantly decreased the proliferation, migration and invasion, but promoted the apoptosis in both cell lines. In addition, R1-P2 treatment effectively inhibited the tumor growth and neovascularization in nude mice with xenografted A549 cells, and R1-P2 also significantly inhibited the FGF2-induced angiogenesis in tube formation experiment and CAM model. We further demonstrated that R1-P2 suppressed lung tumor growth through anti-angiogenic and anti-proliferative activity. Our data may provide a novle leading molecule with potential application in the treatment of FGFR1 activation related lung cancers.


Subject(s)
Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Neovascularization, Pathologic/drug therapy , Peptides/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/metabolism , A549 Cells , Animals , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/metabolism , Peptides/metabolism , Protein Binding , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
14.
J Cell Physiol ; 234(1): 595-605, 2018 01.
Article in English | MEDLINE | ID: mdl-30078186

ABSTRACT

Activation of transforming growth factor-ß (TGF-ß) signaling has been used to enhance healing of meniscal degeneration in several models. However, the exact role and molecular mechanism of TGF-ß signaling in meniscus maintenance and degeneration are still not understood due to the absence of in vivo evidence. In this study, we found that the expression of activin receptor-like kinases 5 (ALK5) in the meniscus was decreased with the progression of age and/or osteoarthritis induced meniscal degeneration. Col2α1 positive cells were found to be specifically distributed in the superficial and inner zones of the anterior horn, as well as the inner zone of the posterior horn in mice, indicating that Col2α1-CreERT2 mice can be a used for studying gene function in menisci. Furthermore, we deleted Alk5 in Col2α1 positive cells in meniscus by administering tamoxifen. Alterations in the menisci structure were evaluated histologically. The expression levels of genes and proteins associated with meniscus homeostasis and TGF-ß signaling were analyzed by quantitative real-time PCR analysis (qRT-PCR) and immunohistochemistry (IHC). Our results revealed severe and progressive meniscal degeneration phenotype in 3- and 6-month-old Alk5 cKO mice compared with Cre-negative control, including aberrantly increased hypertrophic meniscal cells, severe fibrillation, and structure disruption of meniscus. qRT-PCR and IHC results showed that disruption of anabolic and catabolic homeostasis of chondrocytes may contribute to the meniscal degeneration phenotype observed in Alk5 cKO mice. Thus, TGF-ß/ALK5 signaling plays a chondro-protective role in menisci homeostasis, in part, by inhibiting matrix degradation and maintaining extracellular matrix proteins levels in meniscal tissues.


Subject(s)
Collagen Type II/genetics , Meniscus/physiopathology , Osteoarthritis/genetics , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptors, Transforming Growth Factor beta/genetics , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Meniscus/metabolism , Mice , Mice, Knockout , Osteoarthritis/physiopathology , Signal Transduction/genetics , Transforming Growth Factor beta/genetics
15.
EBioMedicine ; 29: 78-91, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29472103

ABSTRACT

p204, a murine member of an interferon-inducible p200 family, was reported to recognize intracellular viral and bacterial DNAs, however, its role in the innate immunity in vivo remains unknown due to the lack of p204-deficient animal models. In this study we first generated the p204-/- mice. Unexpectedly, p204 deficiency led to significant defect in extracellular LPS signaling in macrophages, as demonstrated by dramatic reductions of LPS-mediated IFN-ß and pro-inflammatory cytokines. The serum levels of IFN-ß and pro-inflammatory cytokines were also significantly reduced in p204-/- mice following LPS challenge. In addition, p204-/- mice were resistant to LPS-induced shock. LPS-activated NF-ĸB and IRF-3 pathways were all defective in p204-deficient macrophages. p204 binds to TLR4 through its Pyrin domain, and it is required for the dimerization of TLR4 following LPS-challenge. Collectively, p204 is a critical component of canonical LPS-TLR4 signaling pathway, and these studies also suggest that p204 could be a potential target to prevent and treat inflammatory and infectious diseases.


Subject(s)
Lipopolysaccharides/immunology , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Genotype , Immunity, Innate , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation Mediators/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Macrophages/virology , Mice , Mice, Knockout , Models, Biological , NF-kappa B/metabolism , Nuclear Proteins/genetics , Phosphoproteins/genetics , Protein Binding , Protein Multimerization , RAW 264.7 Cells , Shock, Septic/etiology , Shock, Septic/metabolism , Shock, Septic/mortality , Toll-Like Receptor 4/chemistry
16.
J Bone Miner Res ; 32(11): 2194-2206, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28650109

ABSTRACT

Apert syndrome is one of the most severe craniosynostoses, resulting from gain-of-function mutations in fibroblast growth factor receptor 2 (FGFR2). Previous studies have shown that gain-of-function mutations of FGFR2 (S252W or P253R) cause skull malformation of human Apert syndrome by affecting both chondrogenesis and osteogenesis, underscoring the key role of FGFR2 in bone development. However, the effects of FGFR2 on bone formation at the adult stage have not been fully investigated. To investigate the role of FGFR2 in bone formation, we generated mice with tamoxifen-inducible expression of mutant FGFR2 (P253R) at the adult stage. Mechanical bone marrow ablation (BMX) was performed in both wild-type and Fgfr2 mutant (MT) mice. Changes in newly formed trabecular bone were assessed by micro-computed tomography and bone histomorphometry. We found that MT mice exhibited increased trabecular bone formation and decreased bone resorption after BMX accompanied with a remarkable increase in bone marrow stromal cell recruitment and proliferation, osteoblast proliferation and differentiation, and enhanced Wnt/ß-catenin activity. Furthermore, pharmacologically inhibiting Wnt/ß-catenin signaling can partially reverse the increased trabecular bone formation and decreased bone resorption in MT mice after BMX. Our data demonstrate that gain-of-function mutation in FGFR2 exerts a Wnt/ß-catenin-dependent anabolic effect on trabecular bone by promoting bone formation and inhibiting bone resorption at the adult stage. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Aging/metabolism , Bone Marrow/metabolism , Osteogenesis , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Animals , Bone Resorption/metabolism , Bone Resorption/pathology , Cancellous Bone/metabolism , Cancellous Bone/pathology , Cell Differentiation , Cell Proliferation , Gain of Function Mutation/genetics , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptor, Fibroblast Growth Factor, Type 2/genetics , Up-Regulation , Wnt Signaling Pathway
17.
Sci Rep ; 6: 24042, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-27041213

ABSTRACT

The attenuated degradation of articular cartilage by cartilage-specific deletion of fibroblast growth factor receptor 1 (FGFR1) in adult mice suggests that FGFR1 is a potential target for treating osteoarthritis (OA). The goal of the current study was to investigate the effect of a novel non-ATP-competitive FGFR1 inhibitor, G141, on the catabolic events in human articular chondrocytes and cartilage explants and on the progression of cartilage degradation in a murine model of OA. G141 was screened and identified via cell-free kinase-inhibition assay. In the in vitro study, G141 decreased the mRNA levels of catabolic markers ADAMTS-5 and MMP-13, the phosphorylation of Erk1/2, JNK and p38 MAPK, and the protein level of MMP-13 in human articular chondrocytes. In the ex vivo study, proteoglycan loss was markedly reduced in G141 treated human cartilage explants. For the in vivo study, intra-articular injection of G141 attenuated the surgical destabilization of the medial meniscus (DMM) induced cartilage destruction and chondrocyte hypertrophy and apoptosis in mice. Our data suggest that pharmacologically antagonize FGFR1 using G141 protects articular cartilage from osteoarthritic changes, and intra-articular injection of G141 is potentially an effective therapy to alleviate OA progression.


Subject(s)
Cartilage, Articular/drug effects , Osteoarthritis/drug therapy , Protein Kinase Inhibitors/administration & dosage , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Spiro Compounds/administration & dosage , Animals , Apoptosis , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Injections, Intra-Articular , Mice , Osteoarthritis/genetics , Osteoarthritis/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Spiro Compounds/pharmacology
18.
Autophagy ; 11(11): 1998-2013, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26491898

ABSTRACT

FGFR3 (fibroblast growth factor receptor 3) is a negative regulator of endochondral ossification. Gain-of-function mutations in FGFR3 are responsible for achondroplasia, the most common genetic form of dwarfism in humans. Autophagy, an evolutionarily conserved catabolic process, maintains chondrocyte viability in the growth plate under stress conditions, such as hypoxia and nutritional deficiencies. However, the role of autophagy and its underlying molecular mechanisms in achondroplasia remain elusive. In this study, we found activated FGFR3 signaling inhibited autophagic activity in chondrocytes, both in vivo and in vitro. By employing an embryonic bone culture system, we demonstrated that treatment with autophagy inhibitor 3-MA or chloroquine led to cartilage growth retardation, which mimics the effect of activated-FGFR3 signaling on chondrogenesis. Furthermore, we found that FGFR3 interacted with ATG12-ATG5 conjugate by binding to ATG5. More intriguingly, FGFR3 signaling was found to decrease the protein level of ATG12-ATG5 conjugate. Consistently, using in vitro chondrogenic differentiation assay system, we showed that the ATG12-ATG5 conjugate was essential for the viability and differentiation of chondrocytes. Transient transfection of ATG5 partially rescued FGFR3-mediated inhibition on chondrocyte viability and differentiation. Our findings reveal that FGFR3 inhibits the autophagic activity by decreasing the ATG12-ATG5 conjugate level, which may play an essential role in the pathogenesis of achondroplasia.

19.
Biochim Biophys Acta ; 1843(7): 1237-47, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24657641

ABSTRACT

Fibroblast growth factors (FGFs) and their receptors (FGFRs) play significant roles in vertebrate organogenesis and morphogenesis. FGFR3 is a negative regulator of chondrogenesis and multiple mutations with constitutive activity of FGFR3 result in achondroplasia, one of the most common dwarfisms in humans, but the molecular mechanism remains elusive. In this study, we found that chondrocyte-specific deletion of BMP type I receptor a (Bmpr1a) rescued the bone overgrowth phenotype observed in Fgfr3 deficient mice by reducing chondrocyte differentiation. Consistently, using in vitro chondrogenic differentiation assay system, we demonstrated that FGFR3 inhibited BMPR1a-mediated chondrogenic differentiation. Furthermore, we showed that FGFR3 hyper-activation resulted in impaired BMP signaling in chondrocytes of mouse growth plates. We also found that FGFR3 inhibited BMP-2- or constitutively activated BMPR1-induced phosphorylation of Smads through a mechanism independent of its tyrosine kinase activity. We found that FGFR3 facilitates BMPR1a to degradation through Smurf1-mediated ubiquitination pathway. We demonstrated that down-regulation of BMP signaling by BMPR1 inhibitor dorsomorphin led to the retardation of chondrogenic differentiation, which mimics the effect of FGF-2 on chondrocytes and BMP-2 treatment partially rescued the retarded growth of cultured bone rudiments from thanatophoric dysplasia type II mice. Our findings reveal that FGFR3 promotes the degradation of BMPR1a, which plays an important role in the pathogenesis of FGFR3-related skeletal dysplasia.


Subject(s)
Achondroplasia/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Chondrocytes/metabolism , Growth Plate/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Achondroplasia/metabolism , Achondroplasia/pathology , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Differentiation , Chondrocytes/cytology , Chondrocytes/drug effects , Embryo, Mammalian , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Developmental , Growth Plate/cytology , Growth Plate/growth & development , Humans , Mice , Mice, Knockout , Morphogenesis/genetics , Phosphorylation/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor, Fibroblast Growth Factor, Type 3/deficiency , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
20.
Zhonghua Shao Shang Za Zhi ; 29(4): 338-43, 2013 Aug.
Article in Chinese | MEDLINE | ID: mdl-24351532

ABSTRACT

OBJECTIVE: To study the effects of the panthenol-glutamine on intestinal damage and motor function of intestine in rats with burn injury as well as its dose-effect relationship. METHODS: (1) Experiment 1. Ninety SD rats were divided into groups A-I according to the random number table, with 10 rats in each group. Rats in groups A-I were inflicted with 30% TBSA full-thickness burn and fed by gavage with panthenol and glutamine at post injury hour (PIH) 4, in the whole dosage of 1.00 and 4, 0.50 and 4, 0.25 and 4, 1.00 and 2, 0.50 and 2, 0.25 and 2, 1.00 and 1, 0.50 and 1, 0.25 and 1 g·kg(-1)·d(-1). The feeding was carried out twice a day to achieve the total dosage in 7 days. On drug withdrawal day, blood and intestinal tissue were harvested to detect the intestinal propulsion index, diamine oxidase (DAO) activity in serum, and the content of acetylcholine and intestinal mucosa protein. The best proportion of panthenol and glutamine was screened. (2) Experiment 2. Seventy SD rats were divided into normal control (NC), burn (B), burn+panthenol (B+P), burn+glutamine (B+G), and burn+low, moderate, or high dose of panthenol-glutamine (B+LPG, B+MPG, B+HPG) groups according to the random number table, with 10 rats in each group. Rats in the latter 6 groups were inflicted with 30% TBSA full-thickness burn. Rats in the latter 5 groups were fed by gavage with panthenol and (or) glutamine at PIH 4. Rats in group B+P were fed with panthenol for 1 g·kg(-1)·d(-1), rats in group B+G with glutamine for 4 g·kg(-1)·d(-1), rats in groups B+LPG, B+MPG, and B+HPG with panthenol and glutamine in the dosage of 0.50 and 2, 1.00 and 4, 2.00 and 8 g·kg(-1)·d(-1). The feeding was carried out twice a day to achieve the total dosage for 7 days. The indexes and time point for observation were the same as those of experiment 1. Meanwhile, the pathological change in intestine was observed. The same process was carried out in the rats of group NC. Data were processed with factorial designed analysis of variance (ANOVA), one-way ANOVA and Fisher's exact probability test. LSD was applied for paired comparison. RESULTS: (1) The values of intestinal propulsion index and intestinal mucosa protein content in groups A and B were close (with P values all above 0.05), and were significantly higher than those of the other 7 groups (with P values all below 0.01). Content of acetylcholine in group A was significantly higher than that of the other 8 groups (with P values all below 0.01). DAO activity in groups A, D, and E was close in value (with P values all above 0.05), and all of the values were significantly lower than those of the other 6 groups (with P values all below 0.01). The best proportion of panthenol and glutamine was 1.00 and 4 g·kg(-1)·d(-1). (2) Compared with those of group NC, the intestinal propulsion index, the contents of acetylcholine and intestinal mucosa protein were decreased significantly, while the DAO activity obviously increased in group B (with P values all below 0.01); the intestinal propulsion index was decreased significantly in group B+P (P < 0.01); the intestinal propulsion index and content of acetylcholine were decreased significantly in group B+G (with P values all below 0.01); the intestinal propulsion index was decreased significantly in group B+LPG (P < 0.01); no obvious change was observed in groups B+MPG and B+HPG (with P values all above 0.05). Compared with those of group B [0.50 ± 0.07, (69 ± 10) µg/mL, (26 ± 11) µg/g, (0.672 ± 0.145) U/mL], the contents of acetylcholine and intestinal mucosa protein were increased significantly, DAO activity decreased significantly in group B+P (with P values all below 0.01); the contents of intestinal mucosa protein was increased significantly, DAO activity decreased significantly in group B+G (with P values all below 0.01); the contents of acetylcholine and intestinal mucosa protein were increased significantly in group B+LPG (with P values all below 0.01); the intestinal propulsion index, the contents of acetylcholine and intestinal mucosa protein were increased significantly, while the DAO activity obviously decreased in groups B+MPG and B+HPG [0.66 ± 0.07, 0.68 ± 0.05; (163 ± 24), (168 ± 15) µg/mL; (57 ± 7), (57 ± 7) µg/g; (0.203 ± 0.070), (0.193 ± 0.068) U/mL, with P values all below 0.01]. The levels of the four indexes in groups B+MPG and B+HPG were close or the same in values (with P values all above 0.05). Compared with those of group B, the numbers of rats with irregularly arranged villi in group B+P were decreased significantly (P < 0.05); the numbers of rats with villi decreased in height, irregularly arranged villi, and neutrophil infiltration in group B+G were decreased significantly (with P values all below 0.05); the numbers of rats with villi decreased in height, irregularly arranged villi, degeneration and necrosis of cells, and neutrophil infiltration in group B+LPG were decreased significantly (with P values all below 0.05); the numbers of rats with villi decreased in height and number, irregularly arranged villi, degeneration and necrosis of cells, and neutrophil infiltration in groups B+MPG and B+HPG were decreased significantly (with P values all below 0.05). There was no statistically significant difference between group B+HPG and group B+MPG for the former mentioned five indexes (with P values all above 0.05). CONCLUSIONS: Combined application of panthenol and glutamine can obviously reduce intestinal mucosa damage and promote gastrointestinal motility of rats with burn injury, and they show curative effect superior to exclusive use of either of the two drugs. The best proportion of panthenol and glutamine is 1.00 and 4 g·kg(-1)·d(-1).


Subject(s)
Burns/physiopathology , Glutamine/pharmacology , Intestines/drug effects , Pantothenic Acid/analogs & derivatives , Animals , Dose-Response Relationship, Drug , Female , Gastrointestinal Motility/drug effects , Intestinal Mucosa/drug effects , Intestine, Small , Male , Pantothenic Acid/pharmacology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...