Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Sci Rep ; 14(1): 3291, 2024 02 08.
Article in English | MEDLINE | ID: mdl-38332235

ABSTRACT

Primary human trophoblast stem cells (TSCs) and TSCs derived from human pluripotent stem cells (hPSCs) can potentially model placental processes in vitro. Yet, the pluripotent states and factors involved in the differentiation of hPSCs to TSCs remain poorly understood. In this study, we demonstrate that the primed pluripotent state can generate TSCs by activating pathways such as Epidermal Growth Factor (EGF) and Wingless-related integration site (WNT), and by suppressing tumor growth factor beta (TGFß), histone deacetylases (HDAC), and Rho-associated protein kinase (ROCK) signaling pathways, all without the addition of exogenous Bone morphogenetic protein 4 (BMP4)-a condition we refer to as the TS condition. We characterized this process using temporal single-cell RNA sequencing to compare TS conditions with differentiation protocols involving BMP4 activation alone or BMP4 activation in conjunction with WNT inhibition. The TS condition consistently produced a stable, proliferative cell type that closely mimics first-trimester placental cytotrophoblasts, marked by the activation of endogenous retroviral genes and the absence of amnion expression. This was observed across multiple cell lines, including various primed induced pluripotent stem cell (iPSC) and embryonic stem cell (ESC) lines. Primed-derived TSCs can proliferate for over 30 passages and further specify into multinucleated syncytiotrophoblasts and extravillous trophoblast cells. Our research establishes that the differentiation of primed hPSCs to TSC under TS conditions triggers the induction of TMSB4X, BMP5/7, GATA3, and TFAP2A without progressing through a naive state. These findings propose that the primed hPSC state is part of a continuum of potency with the capacity to differentiate into TSCs through multiple routes.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Humans , Female , Pregnancy , Placenta , Cell Differentiation/genetics , Trophoblasts/metabolism , Bone Morphogenetic Protein 5/metabolism
2.
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
3.
Biochem Biophys Res Commun ; 643: 69-76, 2023 02 05.
Article in English | MEDLINE | ID: mdl-36587524

ABSTRACT

Diabetic peripheral neuropathy is a common and serious complication of diabetes. Bone morphogenetic protein 5 (BMP5) is a multifunctional protein involved in the nervous system. Nevertheless, its effect on diabetic peripheral neuropathy remained uncharacterized. In this study, diabetic neuropathy in mice was induced by a single dose of 150 mg/kg streptozotocin (STZ) via intraperitoneal injection. Lentivirus expressing BMP5 (LV-BMP5) administration improved pain sensitivity, nerve conduction velocities and morphological alterations of the sciatic nerve of diabetic mice. Elevated BMP5 by LV-BMP5 suppressed cell apoptosis in the sciatic nerve, as evidenced by declined TUNEL-positive cells and down-regulated cleaved caspase-3 and cleaved caspase-9 levels. BMP5 enhanced mitochondrial membrane potential and ATP level. BMP5 also increased the phosphorylation of Smad1/5/9. Besides, the role of BMP5 in high glucose (HG)-stimulated Schwann cells was determined. Results of in vitro studies were in line with the in vivo findings. These experimental data seem to imply that BMP5 prevents the development of diabetic neuropathy via the maintenance of Smad1/5/9-mediated mitochondrial function.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Neuropathies , Animals , Mice , Apoptosis , Bone Morphogenetic Protein 5/metabolism , Bone Morphogenetic Protein 5/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diabetic Neuropathies/complications , Mitochondria/metabolism , Schwann Cells/metabolism , Sciatic Nerve/metabolism
4.
J Craniofac Surg ; 33(6): 1934-1938, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35765132

ABSTRACT

ABSTRACT: Bmp5 mutation can lead to microtia in mice models; however, its underlying mechanism is unclear. We analyzed circular RNA (circRNA) expression changes and associated gene regulation during embryonic development of the mouse's external ear after a point mutation occurred naturally in the BMP5 gene. The outer ear tissues of BMP5 short-eared mouse model embryos at embryonic day (E) 15.5 and E17.5 were subjected to RNA sequencing. Changes in the circRNA expression profile were detected using find_circ and the CiRi2 software. Differentially expressed circRNAs were annotated by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses. The circRNA expression profile differed between wild-type and mutant mouse embryos. At E15.5, differentially expressed RNAs were involved in the Hippo signaling pathway, whereas those at E17.5 were associated with stem cell pluripotency. Therefore, circRNA is involved in regulating embryonic external ear development, thus providing a basis for studying the biological aspect of its regulation.


Subject(s)
Bone Morphogenetic Protein 5/metabolism , MicroRNAs , RNA, Circular , Animals , Ear, External , Embryonic Development/genetics , Female , Gene Expression Profiling/methods , Mice , MicroRNAs/genetics , Mutation , Pregnancy , RNA/genetics , RNA, Circular/genetics
5.
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
6.
Prostate ; 81(16): 1435-1449, 2021 12.
Article in English | MEDLINE | ID: mdl-34553788

ABSTRACT

BACKGROUND: Benign prostatic hyperplasia (BPH) is one of the most common illnesses in aging men. Recent studies found that bone morphogenetic protein 5 (BMP5) is upregulated in BPH tissues, however, the role of BMP5 in the development of BPH has not been examined. The current study aims to elucidate the potential roles of BMP5 and related signaling pathways in BPH. METHODS: Human prostate cell lines (BPH-1, WPMY-1) and human/rat hyperplastic prostate tissues were utilized. Western blot, quantitative real-time polymerase chain reaction, immunofluorescent staining, and immunohistochemical staining were performed. BMP5-silenced and -overexpressed cell models were generated and then cell cycle progression, apoptosis, and proliferation were determined. The epithelial-mesenchymal transition (EMT) was also quantitated. And rescue experiments by BMP/Smad signaling pathway agonist or antagonist were accomplished. Moreover, BPH-related tissue microarray analysis was performed and associations between clinical parameters and expression of BMP5 were analyzed. RESULTS: Our study demonstrated that BMP5 was upregulated in human and rat hyperplastic tissues and localized both in the epithelial and stromal compartments of the prostate tissues. E-cadherin was downregulated in hyperplastic tissues, while N-cadherin and vimentin were upregulated. Overexpression of BMP5 enhanced cell proliferation and the EMT process via phosphorylation of Smad1/5/8, while knockdown of BMP5 induced cell cycle arrest at G0/G1 phase and blocked the EMT process. Moreover, a BMP/Smad signaling pathway agonist and antagonist reversed the effects of BMP5 silencing and overexpression, respectively. In addition, BMP5 expression positively correlated with prostate volume and total prostate-specific antigen. CONCLUSION: Our novel data suggest that BMP5 modulated cell proliferation and the EMT process through the BMP/Smad signaling pathway which could contribute to the development of BPH. However, further studies are required to determine the exact mechanism. Our study also indicated that BMP/Smad signaling may be rediscovered as a promising new therapeutic target for the treatment of BPH.


Subject(s)
Bone Morphogenetic Protein 5/metabolism , Epithelial-Mesenchymal Transition/genetics , Prostatic Hyperplasia , Smad Proteins/metabolism , Animals , Apoptosis , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/physiology , Drug Discovery , Gene Knockdown Techniques , Humans , Male , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Rats , Signal Transduction/drug effects , Up-Regulation
7.
Sci Rep ; 11(1): 11978, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099774

ABSTRACT

Anser cygnoides has a spherical crest on the beak roof, which is described as knob. However, the mechanisms affecting knob morphology are unclear. Here, we investigated the phenotypic characteristics and molecular basis of knob-size differences in Yangzhou geese. Anatomically, the knob was identified as frontal hump in the frontal area of the skull, rather than hump of upper beak. Although the frontal hump length, and height varied greatly in geese with different knob phenotypes, little was changed in the width. Histologically, knob skin in large-size knobs geese have a greater length in the stratum corneum, stratum spinosum, and stratum reticular than that in small-size knobs geese. Moveover, the 415 differentially expressed genes were found between the large knobs and small ones through transcriptome profiling. In addition, GO enrichment and KEGG pathway analysis revealed 455 significant GO terms and 210 KEGG pathways were enriched, respectively. Among these, TGF-ß signaling and thyroid hormone synthesis-signaling pathways were identified to determine knob-size phenotype. Furthermore, BMP5, DCN, TSHR and ADCY3 were recognized to involve in the growth and development of knob. Our data provide comprehensive molecular determinants of knob size phenotype, which can potentially promote the genetic improvement of goose knobs.


Subject(s)
Geese/classification , Geese/genetics , Gene Expression Profiling/methods , Adenylyl Cyclases/metabolism , Animals , Base Sequence , Bone Morphogenetic Protein 5/metabolism , Gene Library , Male , Phenotype , Receptors, Thyrotropin/metabolism , Signal Transduction , Skin , Skull , Transcriptome/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.
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
10.
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
11.
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
12.
Nat Commun ; 9(1): 5293, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30546048

ABSTRACT

We used allogeneic bone marrow transplantation (BMT) and a mouse multistage cutaneous carcinogenesis model to probe recruitment of bone marrow-derived epithelial cells (BMDECs) in skin tumors initiated with the carcinogen, dimethylbenz[a]anthracene (DMBA), and promoted with 12-O-tetradecanolyphorbol-13-acetate (TPA). BMDECs clustered in the lesional epithelium, expressed cytokeratins, proliferated, and stratified. We detected cytokeratin induction in plastic-adherent bone marrow cells (BMCs) cultured in the presence of filter-separated keratinocytes (KCs) and bone morphogenetic protein 5 (BMP5). Lineage-depleted BMCs migrated towards High Mobility Group Box 1 (HMGB1) protein and epidermal KCs in ex vivo invasion assays. Naive female mice receiving BMTs from DMBA-treated donors developed benign and malignant lesions after TPA promotion alone. We conclude that BMDECs contribute to the development of papillomas and dysplasia, demonstrating a systemic contribution to these lesions. Furthermore, carcinogen-exposed BMCs can initiate benign and malignant lesions upon tumor promotion. Ultimately, these findings may suggest targets for treatment of non-melanoma skin cancers.


Subject(s)
Bone Marrow Cells/pathology , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Skin Neoplasms/pathology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Bone Marrow Cells/cytology , Bone Marrow Transplantation , Bone Morphogenetic Protein 5/metabolism , Cell Movement , Cell Plasticity/physiology , Coculture Techniques , Epithelial Cells/cytology , Female , HMGB1 Protein/metabolism , Hair Follicle/cytology , Keratinocytes/pathology , Keratins/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness/pathology , Papilloma/pathology , Stem Cells/cytology , Stem Cells/pathology , Tetradecanoylphorbol Acetate/toxicity , Tumor Cells, Cultured
13.
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
14.
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
15.
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
16.
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
17.
Foot Ankle Int ; 37(9): 919-23, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27287343

ABSTRACT

BACKGROUND: Electromagnetic bone growth stimulators have been found to biologically enhance the bone healing environment, with upregulation of numerous growth factors. The purpose of the study was to quantify the effect, in vivo, of pulsed electromagnetic fields (PEMFs) on growth factor expression and healing time in fifth metatarsal nonunions. METHODS: This was a prospective, randomized, double-blind trial of patients, cared for by 2 fellowship-trained orthopedic foot and ankle surgeons. Inclusion criteria consisted of patients between 18 and 75 years old who had been diagnosed with a fifth metatarsal delayed or nonunion, with no progressive signs of healing for a minimum of 3 months. Eight patients met inclusion criteria and were randomized to receive either an active stimulation or placebo PEMF device. Each patient then underwent an open biopsy of the fracture site and was fitted with the appropriate PEMF device. The biopsy was analyzed for messenger-ribonucleic acid (mRNA) levels using quantitative competitive reverse transcription polymerase chain reaction (QT-RT-PCR). Three weeks later, the patient underwent repeat biopsy and open reduction and internal fixation of the nonunion site. The patients were followed at 2- to 4-week intervals with serial radiographs and were graded by the number of cortices of healing. RESULTS: All fractures healed, with an average time to complete radiographic union of 14.7 weeks and 8.9 weeks for the inactive and active PEMF groups, respectively. A significant increase in placental growth factor (PIGF) level was found after active PEMF treatment (P = .043). Other factors trended higher following active PEMF including brain-derived neurotrophic factor (BDNF), bone morphogenetic protein (BMP) -7, and BMP-5. CONCLUSION: The adjunctive use of PEMF for fifth metatarsal fracture nonunions produced a significant increase in local placental growth factor. PEMF also produced trends toward higher levels of multiple other factors and faster average time to radiographic union compared to unstimulated controls. LEVEL OF EVIDENCE: Level I, prospective randomized trial.


Subject(s)
Bone Morphogenetic Protein 5/physiology , Bone Morphogenetic Protein 7/physiology , Brain-Derived Neurotrophic Factor/physiology , Foot Injuries/physiopathology , Fracture Healing/physiology , Fractures, Bone/physiopathology , Metatarsal Bones/physiopathology , Bone Morphogenetic Protein 5/chemistry , Bone Morphogenetic Protein 5/metabolism , Bone Morphogenetic Protein 7/chemistry , Bone Morphogenetic Protein 7/metabolism , Double-Blind Method , Electromagnetic Fields , Humans , Metatarsal Bones/pathology , Metatarsal Bones/physiology , Outcome Assessment, Health Care , Prospective Studies , Transforming Growth Factor beta
18.
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
19.
Drug Des Devel Ther ; 9: 4669-78, 2015.
Article in English | MEDLINE | ID: mdl-26316705

ABSTRACT

Fibroblast activation and proliferation are important for fibroblast-myofibroblast transdifferentiation, a crucial process in the pathological changes that define renal interstitial fibrosis. The left-right determination factor (Lefty) is an important cytokine of the transforming growth factor (TGF)-ß family, with two variants, Lefty-1 and Lefty-2, in mice. Lefty has diverse functions, such as the regulation of embryonic development, the inhibition of TGF-ß1 signaling, and the suppression of tumor activity. However, whether Lefty-1 influences fibroblast activation and proliferation, and consequently prevents fibroblast-myofibroblast transdifferentiation, remains unclear. This study aimed to investigate whether Lefty-1 can attenuate TGF-ß1-induced fibroblast-myofibroblast transdifferentiation in normal rat kidney interstitial fibroblast cells (NRK-49F), as well as the mechanisms underlying any effects. Results showed that the typical fibroblast cell morphology of NRK-49F cells was altered after TGF-ß1 treatment and that Lefty-1 significantly prevented this change in a dose-dependent manner. Further analyses demonstrated decreased proliferating cell nuclear antigen, cyclin D1, collagen I(A1), alpha-smooth muscle actin, and fibronectin expression. Lefty-1 further induced remarkable reductions in TGF-ß1-induced Smad3 and mitogen-activated protein kinase-10/c-Jun N-terminal kinase (JNK-3) signaling, and enhanced expression of the antifibrotic factor bone morphogenetic protein (BMP)-5. However, without TGF-ß1, Lefty-1 had no effect on Smad3, JNK-3, and BMP-5 activation and fibroblast-myofibroblast transdifferentiation. Taken together, these findings indicate that Lefty-1 can alleviate TGF-ß1-mediated activation and the proliferation of fibroblasts. Furthermore, Lefty-1 may prevent fibroblast-myofibroblast transdifferentiation in part via modulations of Smad3, JNK-3, and BMP-5 activities in the TGF-ß/BMP signaling pathway.


Subject(s)
Cell Proliferation/drug effects , Cell Transdifferentiation/drug effects , Fibroblasts/drug effects , Left-Right Determination Factors/pharmacology , Myofibroblasts/drug effects , Transforming Growth Factor beta1/pharmacology , Animals , Bone Morphogenetic Protein 5/metabolism , Cell Line , Cell Shape/drug effects , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Fibrosis , Gene Expression Regulation , Mitogen-Activated Protein Kinase 10/metabolism , Myofibroblasts/metabolism , Rats , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Smad3 Protein/metabolism
20.
Acta Biochim Biophys Sin (Shanghai) ; 47(7): 557-63, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25994008

ABSTRACT

Bone morphogenetic proteins (BMPs) play important roles in tumor cell proliferation, metastasis, and invasion. However, the expression patterns of BMPs in patients with non-small-cell lung cancer (NSCLC) and their correlations with NSCLC pathogenesis have not been examined yet. In this study, the mRNA levels of BMP family members in NSCLC tissues were analyzed and results showed that the mRNA levels of BMP5 and BMP7 were significantly down-regulated and up-regulated, respectively, in tumor tissues compared with those in the corresponding noncancerous tissues. Interestingly, the mRNA level of BMP5 was significantly higher in lung adenocarcinoma tissues than that in lung squamous cell carcinoma tissues. Furthermore, results from immunohistochemistry analysis confirmed stronger expression of BMP5 protein in lung adenocarcinoma than in lung squamous cell carcinoma. Our findings suggested that BMP5 might be a potential prognostic biomarker or therapeutic target for patients with NSCLC.


Subject(s)
Adenocarcinoma/metabolism , Biomarkers, Tumor/metabolism , Bone Morphogenetic Protein 5/metabolism , Carcinoma, Squamous Cell/metabolism , Lung Neoplasms/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL