Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
J Gastroenterol Hepatol ; 39(1): 107-120, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37984826

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is a gastrointestinal tract malignancy. Exosomes secreted by cancer-associated fibroblasts (CAFs) are reported to participate in tumor progression by delivering noncoding RNA or small proteins. However, the function of exosomal miR-522-3p in CRC remains unclear. METHODS: CAFs were derived from tumor tissues, and exosomes were identified by western blot or TEM/NTA and originated from CAFs/NFs. The viability, invasion, and migration of HUVECs and CRC cells was examined using MTT, Transwell, and wound healing assays, respectively. The molecular interactions were validated using dual luciferase reporter assay and RIP. Xenograft and lung metastasis mouse models were generated to assess tumor growth and metastasis. RESULTS: Exosomes extracted from CAFs/NFs showed high expression of CD63, CD81, and TSG101. CAF-derived exosomes significantly increased the viability, angiogenesis, invasion, and migration of HUVECs and CRC cells, thereby aggravating tumor growth, invasion, and angiogenesis in vivo. miR-522-3p was upregulated in CAF-derived exosomes and CRC tissues. Depletion of miR-522-3p reversed the effect of exosomes derived from CAFs in migration, angiogenesis, and invasion of HUVECs and CRC cells. Furthermore, bone morphogenetic protein 5 (BMP5) was identified as a target gene of miR-522-3p, and upregulation of BMP5 reversed the promoting effect of miR-522-3p mimics or CAF-derived exosomes on cell invasion, migration, and angiogenesis of HUVECs and CRC cells. CONCLUSION: Exosomal miR-522-3p from CAFs promoted the growth and metastasis of CRC through downregulating BMP5, which might provide new strategies for the treatment of CRC.


Subject(s)
Cancer-Associated Fibroblasts , Colorectal Neoplasms , Exosomes , MicroRNAs , Animals , Mice , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Bone Morphogenetic Protein 5/genetics , Bone Morphogenetic Protein 5/metabolism , Angiogenesis , Cell Line, Tumor , Exosomes/genetics , Exosomes/metabolism , Colorectal Neoplasms/pathology , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics
2.
J Craniofac Surg ; 34(2): 797-803, 2023.
Article in English | MEDLINE | ID: mdl-36192835

ABSTRACT

This study aimed to perform an association analysis of the full transcriptome in Bmp5 short-ear mice during the development of the external ear in mouse embryos using advanced sequencing techniques. To understand the changes in gene regulation and expression of BMP5 gene mutations involved in the external ear embryonic development of mice, external ear tissues of mouse embryos developed to E15.5 and E17.5 were obtained using a BMP5 short-ear mouse model. The association analysis of the full transcriptome mainly involved the analysis of lncRNA and mRNA associations, the analysis of lncRNA and miRNA associations, the analysis of miRNA and mRNA associations, the analysis of circRNA and mRNA associations and circRNA, miRNA, and mRNA associations. The results showed that regulation of the full transcriptome is associated with external ear development in BMP5 short-ear mouse embryos, and some key regulatory changes in full transcriptome after BMP5 gene point mutation are different. This study will provide a new clue to investigate the mechanism underlying the regulation of mouse external ear development by the full transcriptome.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Mice , Animals , Transcriptome , RNA, Circular , RNA, Long Noncoding/genetics , Mutation , MicroRNAs/genetics , RNA, Messenger/genetics , Embryonic Development , Ear, External , Gene Expression Profiling/methods , Gene Regulatory Networks , Bone Morphogenetic Protein 5/genetics
3.
J Craniofac Surg ; 34(2): 790-796, 2023.
Article in English | MEDLINE | ID: mdl-36166492

ABSTRACT

To understand changes in gene regulation and mRNA expression in external ear development, we used a bone morphogenetic protein 5 (BMP5) short-ear mouse model. External ear tissues at E15.5 and E17.5 were collected, and mRNA expression profiles were analyzed. Upregulated and downregulated mRNA expression was identified using find_circ and CIRI2 software. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed using the differentially expressed mRNAs. Alterations in related signal pathways were identified from the upregulated and downregulated mRNA transcripts. The results showed a correlation between the mRNA expression during external ear development in BMP5 short-ear mice, including key regulatory mRNA changes after point mutations of the Bmp5 gene. This study provides evidence for the mechanism underlying mRNA regulation during external ear development. Changes in mRNA expression profiles also provide clues for future studies regarding the regulatory mechanisms underlying external ear development.


Subject(s)
Ear, External , Gene Expression Regulation , Mice , Animals , Bone Morphogenetic Protein 5/genetics , Mutation , RNA, Messenger/genetics , Gene Expression Profiling/methods , Gene Regulatory Networks
4.
J Craniofac Surg ; 33(8): 2692-2697, 2022.
Article in English | MEDLINE | ID: mdl-35765140

ABSTRACT

ABSTRACT: To understand the changes in gene regulation and expression of MicroRNA (miRNA) involved in external mouseear embryonic development after point mutation of the Bmp5 gene, the outer ear tissues of developed E15.5 and E17.5 mouse embryos were obtained using a Bmp5 short ear mouse model, and the changes in miRNA expression profiles were detected. Changes in miRNA expression in the experimental and control groups were identified during Bmp5 short ear mouse embryo development at E15.5 and E17.5. GO and Kyoto Encyclopedia of Genes and Genomes functional annotations were performed on differentially expressed miRNAs. Multiple signal pathways related to miRNA expression were enhanced during the development of E15.5 and E17.5 embryos of Bmp5 short-ear mice. Based on the basic characteristics of miRNAs, this study aimed to determine the differential expression of miRNAs in Bmp5 short-ear mice during the development of external ear embryos using advanced sequencing techniques. The results showed differences in some key regulatory miRNA changes after point mutations in the Bmp5 gene. This study provides new insights into the mechanism by which miRNAs regulate the development of the external mouse ear. Changes in miRNA expression profiles can also provide clues for studying the biological regulatory mechanism of external ear embryonic development.


Subject(s)
Bone Morphogenetic Protein 5 , Ear, External , Embryonic Development , MicroRNAs , Animals , Mice , Ear, External/embryology , Embryonic Development/genetics , Gene Expression Profiling , Gene Expression Regulation , MicroRNAs/genetics , Mutation , Bone Morphogenetic Protein 5/genetics
5.
Int J Mol Sci ; 23(2)2022 Jan 06.
Article in English | MEDLINE | ID: mdl-35054776

ABSTRACT

Epigenetic abnormalities affect tumor progression, as well as gene expression and function. Among the diverse epigenetic modulators, the histone methyltransferase G9a has been focused on due to its role in accelerating tumorigenesis and metastasis. Although epigenetic dysregulation is closely related to tumor progression, reports regarding the relationship between G9a and its possible downstream factors regulating breast tumor growth are scarce. Therefore, we aimed to verify the role of G9a and its presumable downstream regulators during malignant progression of breast cancer. G9a-depleted MCF7 and T47D breast cancer cells exhibited suppressed motility, including migration and invasion, and an improved response to ionizing radiation. To identify the possible key factors underlying these effects, microarray analysis was performed, and a TGF-ß superfamily member, BMP5, was selected as a prominent target gene. It was found that BMP5 expression was markedly increased by G9a knockdown. Moreover, reduction in the migration/invasion ability of MCF7 and T47D breast cancer cells was induced by BMP5. Interestingly, a G9a-depletion-mediated increase in BMP5 expression induced the phosphorylation of Smad proteins, which are the intracellular signaling mediators of BMP5. Accordingly, we concluded that the observed antitumor effects may be based on the G9a-depletion-mediated increase in BMP5 expression and the consequent facilitation of Smad protein phosphorylation.


Subject(s)
Bone Morphogenetic Protein 5/genetics , Breast Neoplasms/metabolism , Cell Movement , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Neoplasm Invasiveness
6.
Nat Commun ; 13(1): 81, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013172

ABSTRACT

Despite the high prevalence of ischemic heart diseases worldwide, no antibody-based treatment currently exists. Starting from the evidence that a specific isoform of the Bone Morphogenetic Protein 1 (BMP1.3) is particularly elevated in both patients and animal models of myocardial infarction, here we assess whether its inhibition by a specific monoclonal antibody reduces cardiac fibrosis. We find that this treatment reduces collagen deposition and cross-linking, paralleled by enhanced cardiomyocyte survival, both in vivo and in primary cultures of cardiac cells. Mechanistically, we show that the anti-BMP1.3 monoclonal antibody inhibits Transforming Growth Factor ß pathway, thus reducing myofibroblast activation and inducing cardioprotection through BMP5. Collectively, these data support the therapeutic use of anti-BMP1.3 antibodies to prevent cardiomyocyte apoptosis, reduce collagen deposition and preserve cardiac function after ischemia.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bone Morphogenetic Protein 1/genetics , Cardiotonic Agents/pharmacology , Cicatrix/genetics , Endomyocardial Fibrosis/genetics , Myocardial Infarction/genetics , Animals , Bone Morphogenetic Protein 1/antagonists & inhibitors , Bone Morphogenetic Protein 1/metabolism , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 5/genetics , Bone Morphogenetic Protein 5/metabolism , Case-Control Studies , Cell Survival/drug effects , Cicatrix/etiology , Cicatrix/metabolism , Cicatrix/prevention & control , Disease Models, Animal , Endomyocardial Fibrosis/etiology , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/prevention & control , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Troponin T/genetics , Troponin T/metabolism
7.
J Craniofac Surg ; 33(2): e212-e217, 2022.
Article in English | MEDLINE | ID: mdl-34183628

ABSTRACT

ABSTRACT: Microtia is a rare congenital anomaly of the ear; it is regulated by both genetic and environmental factors. However, the mechanisms underlying its pathogenesis are unknown. In this study, the genomes of 2-year-old twin sisters with right microtia were sequenced using human genome-wide sequencing, an approach useful for identifying mutations in genes responsible for congenital microtia. The phenotypes of the twin sisters included congenital microtia on the right side, abnormal auricle shape in the right external ear, a peanut shape for the residual ear, and complete atresia of the right external auditory canal. In the twin sisters, we identified a previously unknown mutation in BMP5(exon4:c.833- 4C>G), as well as a new mutation (exon2:c.G332T:p.S111I) in BMP2, both of which were confirmed using polymerase chain reaction-based amplification of the corresponding genome regions, followed by first-generation sequencing. The exon4:c.833-4C>G mutation in human BMP5 may be the main cause of microtia in the twin sisters. A pathogenic mutation in human BMP2 (exon2:c.G332T:p.S111I) may be responsible for the facial deformity in the twin sisters. Thus, our study demonstrates the potential of genome-wide sequencing for identifying novel mutations associated with microtia on the whole-genome scale and extends the mutation spectrum of BMP5. Additionally, our data suggest that BMP2 is another pathogenic gene associated with microtia.


Subject(s)
Congenital Microtia , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 5/genetics , Congenital Microtia/genetics , Ear Canal/abnormalities , Humans , Mutation , Phenotype , Twins, Monozygotic/genetics
8.
Aging (Albany NY) ; 13(7): 9646-9664, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33744859

ABSTRACT

In this study, we using the in vivo destabilization of the medial meniscus (DMM) mouse model to investigate the role of bone morphogenetic protein 5 (BMP5) in osteoarthritis (OA) progression mediated via chondrocyte senescence and apoptosis. BMP5 expression was significantly higher in knee articular cartilage tissues of OA patients and DMM model mice than the corresponding controls. The Osteoarthritis Research Society International scores based on histological staining of knee articular cartilage sections were lower in DMM mice where BMP5 was knocked down in chondrocytes than the corresponding controls 4 weeks after DMM surgery. DMM mice with BMP5-deficient chondrocytes showed reduced levels of matrix-degrading enzymes such as MMP13 and ADAMTS5 as well as reduced cartilage destruction. BMP5 knockdown also decreased chondrocyte apoptosis and senescence by suppressing the activation of p38 and ERK MAP kinases. These findings demonstrate that BMP5 silencing inhibits chondrocyte senescence and apoptosis as well as OA progression by downregulating activity in the p38/ERK signaling pathway.


Subject(s)
Apoptosis/physiology , Bone Morphogenetic Protein 5/metabolism , Cellular Senescence/physiology , Chondrocytes/metabolism , Osteoarthritis/metabolism , ADAMTS5 Protein/genetics , ADAMTS5 Protein/metabolism , Animals , Bone Morphogenetic Protein 5/genetics , Cartilage, Articular/metabolism , Cell Line , Disease Progression , Gene Silencing , Humans , MAP Kinase Signaling System/physiology , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Mice , Osteoarthritis/genetics
9.
Sci Rep ; 10(1): 15579, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32968094

ABSTRACT

Barrett's esophagus (BE) predisposes for the malignant condition of esophageal adenocarcinoma (EAC). Since BE patients have few or no symptoms, most of these patients are not identified and not included in surveillance programs. These BE patients are at risk of developing advanced-stage EAC. At present, non-invasive tests to identify BE patients from the general population are lacking. We and others showed that Bone Morphogenetic Protein 4 (BMP4), and other BMPs are upregulated in BE. We aimed to determine if circulating BMPs can be identified and used as blood biomarkers to identify BE patients at high risk in the general population. In this study, we could detect the different BMPs in the blood of 112 BE patients and 134 age- and sex-matched controls. Concentration levels of BMP2, BMP4, and BMP5 were elevated in BE patients, with BMP2 and BMP5 significantly increased. BMP5 remained significant after multivariate analysis and was associated with an increased risk for BE with an OR of 1.49 (p value 0.01). Per log (pg/mL) of BMP5, the odds of having BE increased by 50%. Future optimization and validation studies might be needed to prove its utility as a non-invasive method for the detection of BE in high-risk populations and screening programs.


Subject(s)
Barrett Esophagus/blood , Biomarkers/blood , Bone Morphogenetic Protein 5/blood , Aged , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Bone Morphogenetic Protein 2/blood , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 4/blood , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 5/genetics , Female , Gene Expression Regulation/genetics , Humans , Male , Middle Aged , Risk Factors
10.
Elife ; 92020 09 07.
Article in English | MEDLINE | ID: mdl-32894216

ABSTRACT

Tissue homeostasis relies on the fine regulation between stem and progenitor cell maintenance and lineage commitment. In the adult prostate, stem cells have been identified in both basal and luminal cell compartments. However, basal stem/progenitor cell homeostasis is still poorly understood. We show that basal stem/progenitor cell maintenance is regulated by a balance between BMP5 self-renewal signal and GATA3 dampening activity. Deleting Gata3 enhances adult prostate stem/progenitor cells self-renewal capacity in both organoid and allograft assays. This phenotype results from a local increase in BMP5 activity in basal cells as shown by the impaired self-renewal capacity of Bmp5-deficient stem/progenitor cells. Strikingly, Bmp5 gene inactivation or BMP signaling inhibition with a small molecule inhibitor are also sufficient to delay prostate and skin cancer initiation of Pten-deficient mice. Together, these results establish BMP5 as a key regulator of basal prostate stem cell homeostasis and identifies a potential therapeutic approach against Pten-deficient cancers.


Subject(s)
Bone Morphogenetic Protein 5 , Prostate/metabolism , Prostatic Neoplasms , Stem Cells/metabolism , Animals , Bone Morphogenetic Protein 5/genetics , Bone Morphogenetic Protein 5/metabolism , Homeostasis , Male , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism
11.
JCI Insight ; 52019 05 16.
Article in English | MEDLINE | ID: mdl-31094703

ABSTRACT

Benign prostatic hyperplasia (BPH) is the most common cause of lower urinary tract symptoms in men. Current treatments target prostate physiology rather than BPH pathophysiology and are only partially effective. Here, we applied next-generation sequencing to gain new insight into BPH. By RNAseq, we uncovered transcriptional heterogeneity among BPH cases, where a 65-gene BPH stromal signature correlated with symptom severity. Stromal signaling molecules BMP5 and CXCL13 were enriched in BPH while estrogen regulated pathways were depleted. Notably, BMP5 addition to cultured prostatic myofibroblasts altered their expression profile towards a BPH profile that included the BPH stromal signature. RNAseq also suggested an altered cellular milieu in BPH, which we verified by immunohistochemistry and single-cell RNAseq. In particular, BPH tissues exhibited enrichment of myofibroblast subsets, whilst depletion of neuroendocrine cells and an estrogen receptor (ESR1)-positive fibroblast cell type residing near epithelium. By whole-exome sequencing, we uncovered somatic single-nucleotide variants (SNVs) in BPH, of uncertain pathogenic significance but indicative of clonal cell expansions. Thus, genomic characterization of BPH has identified a clinically-relevant stromal signature and new candidate disease pathways (including a likely role for BMP5 signaling), and reveals BPH to be not merely a hyperplasia, but rather a fundamental re-landscaping of cell types.


Subject(s)
Genetic Predisposition to Disease/genetics , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Bone Morphogenetic Protein 5/genetics , Bone Morphogenetic Protein 5/metabolism , Exome , Humans , Male , Myofibroblasts , Neuroendocrine Cells , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Estrogen , Severity of Illness Index , Transcriptome
12.
Cell Death Dis ; 10(4): 272, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894517

ABSTRACT

Several studies have reported inducing adult cells into sweat gland-like cells; however, slow transition and low efficiency limit the potential for cell-based treatment. Here, we show that overexpression of the transcription factor FoxC1 was sufficient to reprogram epidermal cells to induced functional sweat gland-like cells (iSGCs). The iSGCs expressing secreting-related genes, had a global gene expression profile between fetal SGCs (P5) and adult SGCs (P28). Moreover, iSGCs transplanted into the burn mice model facilitated wound repair and sweat gland regeneration. We further demonstrated that the Foxc1 upregulated BMP5 transcription and BMP5 is responsible for the cell-type transition. Collectively, this study shows that lineage reprogramming of epidermal cells into iSGCs provides an excellent cell source and a promising regenerative strategy for anhidrosis and hypohidrosis.


Subject(s)
Cellular Reprogramming/genetics , Epidermal Cells/metabolism , Forkhead Transcription Factors/metabolism , Sweat Glands/cytology , Animals , Bone Morphogenetic Protein 5/genetics , Bone Morphogenetic Protein 5/metabolism , Burns/metabolism , Burns/therapy , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Transplantation/methods , Forkhead Transcription Factors/genetics , Gene Knockdown Techniques , Hypohidrosis/therapy , Interferon Regulatory Factors/metabolism , Mice , Mice, Inbred C57BL , Repressor Proteins/metabolism , Transcriptome , Transfection , Wound Healing/physiology
13.
Mol Cancer ; 17(1): 176, 2018 12 20.
Article in English | MEDLINE | ID: mdl-30572883

ABSTRACT

BACKGROUND: Although the genetic spectrum of human colorectal cancer (CRC) is mainly characterized by APC, KRAS and TP53 mutations, driver genes in tumor initiation have not been conclusively demonstrated. In this study, we aimed to identify novel markers for CRC. METHODS: We performed exome analysis of sporadic colorectal cancer (sCRC) coding regions to screen loss of function (LoF) mutation genes, and carried out systems-level approaches to confirm top rank gene in this study. RESULTS: We identified loss of BMP5 is an early event in CRC. Deep sequencing identified BMP5 was mutated in 7.7% (8/104) of sCRC samples, with 37.5% truncating mutation frequency. Notably, BMP5 negative expression and its prognostic value is uniquely significant in sCRC but not in other tumor types. Furthermore, BMP5 expression was positively correlated with E-cadherin in CRC patients and its dysregulation play a vital role in epithelial-mesenchymal transition (EMT), thus triggering tumor initiation and development. RNA sequencing identified, independent of BMP/Smads pathway, BMP5 signaled though Jak-Stat pathways to inhibit the activation of oncogene EPSTI1. CONCLUSIONS: Our result support a novel concept that the importance of BMP5 in sCRC. The tumor suppressor role of BMP5 highlights its crucial role in CRC initiation and development.


Subject(s)
Bone Morphogenetic Protein 5/genetics , Colorectal Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling/methods , HCT116 Cells , HT29 Cells , Humans , Mutation/genetics , Signal Transduction , Smad Proteins/genetics , Transcriptome
14.
Cell Syst ; 7(4): 422-437.e7, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30268436

ABSTRACT

We present an integromic analysis of gene alterations that modulate transforming growth factor ß (TGF-ß)-Smad-mediated signaling in 9,125 tumor samples across 33 cancer types in The Cancer Genome Atlas (TCGA). Focusing on genes that encode mediators and regulators of TGF-ß signaling, we found at least one genomic alteration (mutation, homozygous deletion, or amplification) in 39% of samples, with highest frequencies in gastrointestinal cancers. We identified mutation hotspots in genes that encode TGF-ß ligands (BMP5), receptors (TGFBR2, AVCR2A, and BMPR2), and Smads (SMAD2 and SMAD4). Alterations in the TGF-ß superfamily correlated positively with expression of metastasis-associated genes and with decreased survival. Correlation analyses showed the contributions of mutation, amplification, deletion, DNA methylation, and miRNA expression to transcriptional activity of TGF-ß signaling in each cancer type. This study provides a broad molecular perspective relevant for future functional and therapeutic studies of the diverse cancer pathways mediated by the TGF-ß superfamily.


Subject(s)
Mutation Rate , Neoplasms/genetics , Signal Transduction , Transforming Growth Factor beta/metabolism , Bone Morphogenetic Protein 5/genetics , Bone Morphogenetic Protein 5/metabolism , DNA Methylation , Humans , MicroRNAs/genetics , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta/genetics
15.
Biomed Pharmacother ; 106: 1046-1051, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30119170

ABSTRACT

MiRNA regulation is a crucial way of epigenetic changes. Mir-32 has been reported in several studies as an oncogene, however, its role and target in colorectal cancer (CRC) remains unclear. In this study, we aimed to explore the role of miR-32 in CRC using bioinformatic analysis and functional assays. We collected 28 pairs of CRC tumor tissues and adjacent normal tissues and confirmed miR-32 was significantly upregulated in CRC. Expression and clinical data from The Cancer Genome Atlas (TCGA) identified miR-32 expression is associated with CRC lymphatic invasion, metastasis, and correlates with patients' poor survival. Functional studies demonstrated that overexpression of miR-32 in LoVo cells promoted cell proliferation and migration, whereas inhibition of miR-32 in HCT 116 cells showed the opposite results. Using bioinformatics, we identified Bone morphogenetic protein 5 (BMP5) is a direct target of miR-32, and loss of tumor suppressor BMP5 may partially due to the miR-32 dysregulation. The inverse correlation between miR-32 and BMP5 was observed in CRC, especially in advanced tumor patients. Moreover, cotransfection of miR-32 mimics and BMP5 recombinant vector in LoVo cells demonstrated that BMP5 could reverse the oncomir effect of miR-32. Taken together, our results suggested a significant role of miR-32/BMP5 axis in CRC tumorigenesis.


Subject(s)
Bone Morphogenetic Protein 5/metabolism , Colorectal Neoplasms/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Binding Sites , Bone Morphogenetic Protein 5/genetics , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Computational Biology , Databases, Genetic , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis , MicroRNAs/genetics , Neoplasm Invasiveness , Signal Transduction , Time Factors
16.
J Neurosci ; 38(7): 1662-1676, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29321139

ABSTRACT

The embryonic formation of midbrain dopaminergic (mDA) neurons in vivo provides critical guidelines for the in vitro differentiation of mDA neurons from stem cells, which are currently being developed for Parkinson's disease cell replacement therapy. Bone morphogenetic protein (BMP)/SMAD inhibition is routinely used during early steps of stem cell differentiation protocols, including for the generation of mDA neurons. However, the function of the BMP/SMAD pathway for in vivo specification of mammalian mDA neurons is virtually unknown. Here, we report that BMP5/7-deficient mice (Bmp5-/-; Bmp7-/-) lack mDA neurons due to reduced neurogenesis in the mDA progenitor domain. As molecular mechanisms accounting for these alterations in Bmp5-/-; Bmp7-/- mutants, we have identified expression changes of the BMP/SMAD target genes MSX1/2 (msh homeobox 1/2) and SHH (sonic hedgehog). Conditionally inactivating SMAD1 in neural stem cells of mice in vivo (Smad1Nes) hampered the differentiation of progenitor cells into mDA neurons by preventing cell cycle exit, especially of TH+SOX6+ (tyrosine hydroxylase, SRY-box 6) and TH+GIRK2+ (potassium voltage-gated channel subfamily-J member-6) substantia nigra neurons. BMP5/7 robustly increased the in vitro differentiation of human induced pluripotent stem cells and induced neural stem cells to mDA neurons by up to threefold. In conclusion, we have identified BMP/SMAD signaling as a novel critical pathway orchestrating essential steps of mammalian mDA neurogenesis in vivo that balances progenitor proliferation and differentiation. Moreover, we demonstrate the potential of BMPs to improve the generation of stem-cell-derived mDA neurons in vitro, highlighting the importance of sequential BMP/SMAD inhibition and activation in this process.SIGNIFICANCE STATEMENT We identify bone morphogenetic protein (BMP)/SMAD signaling as a novel essential pathway regulating the development of mammalian midbrain dopaminergic (mDA) neurons in vivo and provide insights into the molecular mechanisms of this process. BMP5/7 regulate MSX1/2 (msh homeobox 1/2) and SHH (sonic hedgehog) expression to direct mDA neurogenesis. Moreover, the BMP signaling component SMAD1 controls the differentiation of mDA progenitors, particularly to substantia nigra neurons, by directing their cell cycle exit. Importantly, BMP5/7 increase robustly the differentiation of human induced pluripotent and induced neural stem cells to mDA neurons. BMP/SMAD are routinely inhibited in initial stages of stem cell differentiation protocols currently being developed for Parkinson's disease cell replacement therapies. Therefore, our findings on opposing roles of the BMP/SMAD pathway during in vitro mDA neurogenesis might improve these procedures significantly.


Subject(s)
Bone Morphogenetic Proteins/physiology , Dopaminergic Neurons/physiology , Mesencephalon/physiology , Neural Stem Cells , Neurogenesis/physiology , Pluripotent Stem Cells , Signal Transduction/physiology , Smad Proteins/physiology , Animals , Bone Morphogenetic Protein 5/genetics , Bone Morphogenetic Protein 5/metabolism , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Cell Proliferation , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , Mesencephalon/cytology , Mice , Mice, Knockout , Smad1 Protein/genetics , Smad1 Protein/metabolism
17.
J Postgrad Med ; 63(3): 151-156, 2017.
Article in English | MEDLINE | ID: mdl-28695869

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is a chronic degenerative disorder probably affected by both genetic and environmental causes. Bone morphogenetic proteins (BMPs) are bone-derived factors that can induce new bone formation. Single-nucleotide polymorphisms (SNPs) of BMP5 gene alters the transcriptional activity of the BMP5 promoter that has been involved in OA susceptibility. This case-control study investigated the association of rs1470527 and rs9382564 SNP of BMP5 gene with susceptibility to knee OA (KOA). MATERIALS AND METHODS: A total of 499 cases with radiographic KOA and 458 age- and sex-matched healthy controls were enrolled. Venous blood samples were obtained from all the cases as well as controls for polymerase chain reaction-restriction fragment length polymorphism. RESULTS: The genotype distribution for rs1470527 and rs9382564 SNP was significantly different in cases and controls (P < 0.0001). Within both the SNPs of BMP5 gene, genotype CT and TT were significantly (P < 0.0001) associated with KOA as compared to the CC genotype. T allele of both the studied SNP was significantly associated with KOA (P < 0.0001). The allele frequencies of rs1470527 were 0.56(T) and 0.44(C) in cases and 0.33(T) and 0.67(C) in controls and in rs9382564 were 0.57(C) and 0.43(T) in cases and 0.71(C) and 0.29(T) in controls. Further in relation with clinical severity of OA, we observed signification association of TT genotype with both visual analog scale (P < 0.0001) and Western Ontario and McMaster Universities score (P < 0.05). CONCLUSION: Our results indicate significant association of rs1470527 and rs9382564 polymorphism of BMP5 gene with KOA.


Subject(s)
Asian People/genetics , Bone Morphogenetic Protein 5/genetics , Osteoarthritis, Knee/diagnostic imaging , Adult , Aged , Case-Control Studies , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Osteoarthritis, Knee/genetics , Polymorphism, Restriction Fragment Length , Polymorphism, Single Nucleotide
18.
Biofactors ; 43(4): 558-566, 2017 Jul 08.
Article in English | MEDLINE | ID: mdl-28497879

ABSTRACT

Bone morphogenetic protein-5 (BMP-5) is a member of the TGF receptor-ß family with osteoinductive property. However, its physiological role in osteoblast differentiation is not defined. This study highlights the importance of BMP-5 in MC3T3E1 osteoblast differentiation. Pre-osteoblasts exposed to osteogenic media (ascorbic acid, 50 µg/ml and ß-glycerophosphate, 10 mM) showed high protein expression of BMP-5 in cell lysates and cell culture supernatants, which peaked during early time-points of differentiation and declined with onset of mineralization. Attenuation of endogenous BMP-5 protein expression by RNA interference downregulated the expression of type I collagen (COLIA1), an early osteoblast differentiation marker but not osteocalcin, a late osteoblast differentiation marker. Further experiments to analyze the cell signaling components revealed that BMP-5 modulates COLIA1 expression via p38-Runx2 axis involving Runx2 (Ser19) phosphorylation. These effects were also observed when recombinant BMP-5 was added to pre-osteoblast cultures reinforcing the fact that BMP-5 is a modulator of COLIA1 expression. We conclude that BMP-5 has stage-specific role to play during MC3T3E1 osteoblast differentiation in part by autocrine p38/Runx2/COLIA1 signaling. © 2017 BioFactors, 43(4):558-566, 2017.


Subject(s)
Bone Morphogenetic Protein 5/metabolism , Osteoblasts/metabolism , Animals , Bone Morphogenetic Protein 5/genetics , Cell Differentiation , Cell Line , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Enzyme-Linked Immunosorbent Assay , Mice , Microscopy, Electron, Scanning , Phosphorylation , RNA, Small Interfering , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Mol Cancer ; 16(1): 74, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28372542

ABSTRACT

BACKGROUND: The telomerase/telomere interacting protein PinX1 has been suggested as a tumor suppressor. However, the clinical and biological significance of PinX1 in human non-small cell lung cancer (NSCLC) is unclear. METHODS: PinX1 gene/expression pattern and its association with NSCLC patient survival were analyzed in cBioportal Web resource and two cohorts of NSCLC samples. A series of in vivo and in vitro assays were performed to elucidate the function of PinX1 on NSCLC cells proliferation and underlying mechanisms. RESULTS: More frequency of gene PinX1 homozygous deletion and heterozygote deficiency was first retrieved from cBioportal Web resource. Low expression of PinX1 correlated with smoking condition, histological type, T stage, N stage, M stage and TNM stage, and was an independent predictor for overall survival in a learning cohort (n = 93) and a validation cohort (n = 51) of NSCLC patients. Furthermore, knockdown of PinX1 dramatically accelerated NSCLC cell proliferation and G1/S transition, whereas ectopic overexpression of PinX1 substantially inhibited cell viability and cell cycle transition in vitro and in vivo. p15/cyclin D1 pathway and BMP5 might contribute to PinX1-associated cell proliferation and cell cycle transition. CONCLUSION: The cost-effective expression of PinX1 could constitute a novel molecular predictor/marker for NSCLC management.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics , Adult , Aged , Animals , Biomarkers, Tumor , Bone Morphogenetic Protein 5/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Cell Cycle , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Databases, Nucleic Acid , Disease Models, Animal , Female , Gene Deletion , Gene Silencing , Humans , Lung Neoplasms/mortality , Male , Mice , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Xenograft Model Antitumor Assays
20.
J Cell Biochem ; 117(11): 2658-68, 2016 11.
Article in English | MEDLINE | ID: mdl-27038365

ABSTRACT

The in vitro osteogenic differentiation has been intensively studied. However, it is not yet clear precisely how osteogenesis can be optimized. Changes in extracellular Ca(2+) concentration ([Ca(2+) ]e ), as well as modulation of purinergic receptors play an important role in the regulation of osteoblasts differentiation and bone formation. In this study, we investigated the effects of a combined treatment of ATPγ-S and high [Ca(2+) ]e (5.35 mM) on osteogenic differentiation and function of primary cell cultures from rat calvaria. Our results indicate that ATPγ-S stimulates cell transition from the G0 to S phase of cell cycle, involving the PI3K signaling pathway. Treatment with 10 or 100 µM ATPγ-S and [Ca(2+) ]e (ATP-[Ca(2+) ]e ) for 48 h increases cell number significantly above the control. ATPγ-S treatment in osteogenic medium containing [Ca(2+) ]e stimulates the gene expression of BMP-4, BMP-5, and OPN at 16, 48, and 72 h, respectively, above control. In same conditions, treatment for 6 days with 10 µM UTP or 100 µM UDP significantly increased the ALP activity respect to control. Cells grown in osteogenic medium showed a statistically significant increase in calcium deposits at 15 and 18 days, for 10 µM ATPγ-S treatment, and at 18 and 22 days, for [Ca(2+) ]e treatment, respect to control but ATP-[Ca(2+) ]e treatment shown a significant greater mineralization at 15 days respect to ATPγ-S, and at 18 days respect to both agonists. In conclusion, we demonstrated that an osteogenic medium containing 10 µM ATPγ-S and 5.35 mM [Ca(2+) ]e enhance osteogenesis and mineralization by rat primary calvarial cells cultures. J. Cell. Biochem. 117: 2658-2668, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 5/metabolism , Calcium/pharmacology , Cell Differentiation/drug effects , Cytoskeletal Proteins/metabolism , GTPase-Activating Proteins/metabolism , Nuclear Proteins/metabolism , Osteogenesis/physiology , Skull/cytology , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Blotting, Western , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 5/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cytoskeletal Proteins/genetics , Drug Combinations , GTPase-Activating Proteins/genetics , Nuclear Proteins/genetics , Osteogenesis/drug effects , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skull/drug effects , Skull/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...