Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
J Orthop Res ; 41(3): 511-523, 2023 03.
Article in English | MEDLINE | ID: mdl-35716158

ABSTRACT

Entheses, which are tendon-to-bone attachment sites in the musculoskeletal system, play important roles in optimizing the mechanical stress and force transmitted from the muscle to the bone. Sports-related enthesopathy shows pathological features, including hyperplasia of the fibrocartilage (FC) region in the enthesis. The amount of exercise and type of muscle contraction during movement is involved in the pathogenesis of sports-related enthesopathy; however, the details of this condition are unclear. Here we examined the molecular pathways involved in the morphological changes of the muscle-tendon-enthesis complex and enthesis FC region in the supraspinatus muscle enthesis of mice under different exercise conditions. Following intervention, morphological changes in the muscle-tendon-enthesis complex were initiated in the eccentric contraction-dominant exercise group at 2 weeks, with activation of the transforming growth factor-ß (TGFß) superfamily pathway predicted by proteome and ingenuity pathway analyses. Histological and molecular biological analyses confirmed the activation of the TGFß/bone morphogenetic protein (BMP)-Smad pathway. The concentric contraction-dominant exercise group showed no change in the morphology of the muscle-tendon-enthesis complex or activation of the TGFß/BMP-Smad pathway, despite overuse exercise. Statement of Clinical Significance: These results suggest that eccentric contraction-dominant exercise induces sports-related enthesopathy-like morphological changes in the early stages as well as molecular biological changes, mainly in the transforming growth factor-ß superfamily pathway in enthesis. Statement of Clinical Significance: These results suggest that eccentric contraction-dominant exercise induces sports-related enthesopathy-like morphological changes in the early stages as well as molecular biological changes, mainly in the transforming growth factor-ß superfamily pathway in enthesis.


Subject(s)
Enthesopathy , Physical Conditioning, Animal , TGF-beta Superfamily Proteins , Animals , Mice , Bone and Bones/pathology , Tendons/pathology , TGF-beta Superfamily Proteins/metabolism
2.
Methods Mol Biol ; 2488: 67-80, 2022.
Article in English | MEDLINE | ID: mdl-35347683

ABSTRACT

Transforming growth factor-ß (TGF-ß) family members have pivotal functions in controlling breast cancer progression, acting not only on cancer cells but also on other cells within the tumor microenvironment. Here we describe embryonic zebrafish xenograft assays to investigate how TGF-ß family signaling controls breast cancer cell intravasation, extravasation and regulates tumor angiogenesis. Fluorescently mCherry-labeled breast cancer cells are injected in the perivitelline space or Duct of Cuvier of Tg (fli:EGFP) transgenic Casper zebrafish embryos, in which the zebrafish express enhanced green fluorescent protein in the entire vasculature. The dynamic responses of migratory and invasive human cancer cells, and the induction of new blood vessel formation by the cancer cells in zebrafish host, are visualized using a fluorescent microscope. These assays provide efficient, reliable, low-cost models to investigate the effect of (epi)genetic modulators and pharmacological compounds that perturb the activity of TGF-ß family signaling components on breast cancer cell metastasis and angiogenesis.


Subject(s)
Breast Neoplasms , TGF-beta Superfamily Proteins/metabolism , Zebrafish , Animals , Breast Neoplasms/pathology , Female , Heterografts , Humans , Neoplasm Transplantation , Signal Transduction , Tumor Microenvironment , Zebrafish/metabolism
3.
J Mol Biol ; 434(5): 167439, 2022 03 15.
Article in English | MEDLINE | ID: mdl-34990654

ABSTRACT

The 33 members of the transforming growth factor beta (TGF-ß) family are fundamentally important for organismal development and homeostasis. Family members are synthesized and secreted as pro-complexes of non-covalently associated prodomains and growth factors (GF). Pro-complexes from a subset of family members are latent and require activation steps to release the GF for signaling. Why some members are latent while others are non-latent is incompletely understood, particularly because of large family diversity. Here, we have examined representative family members in negative stain electron microscopy (nsEM) and hydrogen deuterium exchange (HDX) to identify features that differentiate latent from non-latent members. nsEM showed three overall pro-complex conformations that differed in prodomain arm domain orientation relative to the bound growth factor. Two cross-armed members, TGF-ß1 and TGF-ß2, were each latent. However, among V-armed members, GDF8 was latent whereas ActA was not. All open-armed members, BMP7, BMP9, and BMP10, were non-latent. Family members exhibited remarkably varying HDX patterns, consistent with large prodomain sequence divergence. A strong correlation emerged between latency and protection of the prodomain α1-helix from exchange. Furthermore, latency and protection from exchange correlated structurally with increased α1-helix buried surface area, hydrogen bonds, and cation-pi bonds. Moreover, a specific pattern of conserved basic and hydrophobic residues in the α1-helix and aromatic residues in the interacting fastener were found only in latent members. Thus, this first comparative survey of TGF-ß family members reveals not only diversity in conformation and dynamics but also unique features that distinguish latent members.


Subject(s)
TGF-beta Superfamily Proteins , Hydrogen Bonding , Protein Conformation, alpha-Helical , Protein Domains , Signal Transduction , TGF-beta Superfamily Proteins/chemistry , TGF-beta Superfamily Proteins/metabolism
4.
Zygote ; 30(1): 65-71, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33966679

ABSTRACT

Regulation of the transforming growth factor beta (TGFß) superfamily by gonadotrophins in swine follicular cells is not fully understood. This study evaluated the expression of steroidogenic enzymes and members of the TGFß superfamily in prepubertal gilts allocated to three treatments: 1200 IU eCG at D -3 (eCG); 1200 IU eCG at D -6 plus 500 IU hCG at D -3 (eCG + hCG); and the control, composed of untreated gilts. Blood samples and ovaries were collected at slaughter (D0) and follicular cells were recovered thereafter. Relative gene expression was determined by real-time PCR. Serum progesterone levels were greater in the eCG + hCG group compared with the other groups (P < 0.01). No differences were observed in the expression of BMP15, BMPR1A, BMPR2, FSHR, GDF9, LHCGR and TGFBR1 (P > 0.05). Gilts from the eCG group presented numerically greater mean expression of CYP11A1 mRNA than in the control group that approached statistical significance (P = 0.08) and greater expression of CYP19A1 than in both the eCG and the control groups (P < 0.05). Expression of BMPR1B was lower in the eCG + hCG treatment group compared with the control (P < 0.05). In conclusion, eCG treatment increased the relative expression of steroidogenic enzymes, whereas treatment with eCG + hCG increased serum progesterone levels. Although most of the evaluated TGFß members were not regulated after gonadotrophin treatment, the downregulation of BMPR1B observed after treatment with eCG + hCG and suggests a role in luteinization regulation.


Subject(s)
Chorionic Gonadotropin , Ovarian Follicle/cytology , TGF-beta Superfamily Proteins/metabolism , Animals , Chorionic Gonadotropin/pharmacology , Female , Progesterone , Swine
5.
Pediatr Rheumatol Online J ; 19(1): 72, 2021 May 12.
Article in English | MEDLINE | ID: mdl-33980237

ABSTRACT

BACKGROUND: We examined influences of conditioned media from chondrocytes (Ch) on juvenile idiopathic arthritis synovial fibroblasts (JFLS) and potential for JFLS to undergo endochondral bone formation (EBF). METHODS: Primary cells from three control fibroblast-like synoviocytes (CFLS) and three JFLS were cultured in Ch-conditioned media and compared with untreated fibroblast-like synoviocytes (FLS). RNA was analyzed by ClariomS microarray. FLS cells cultured in conditioned media were exposed to either TGFBR1 inhibitor LY3200882 or exogenous BMP4 and compared with FLS cultured in conditioned media from Ch (JFLS-Ch). Media supernatants were analyzed by ELISA. RESULTS: In culture, JFLS downregulate BMP2 and its receptor BMPR1a while upregulating BMP antagonists (NOG and CHRD) and express genes (MMP9, PCNA, MMP12) and proteins (COL2, COLX, COMP) associated with chondrocytes. Important TGFß superfamily member gene expression (TGFBI, MMP9, COL1A1, SOX6, and MMP2) is downregulated when JFLS are cultured in Ch-conditioned media. COL2, COLX and COMP protein expression decreases in JFLS-Ch. BMP antagonist protein (NOG, CHRD, GREM, and FST) secretion is significantly increased in JFLS-Ch. Protein phosphorylation increases in JFLS-Ch exposed to exogenous BMP4, and chondrocyte-like phenotype is restored in BMP4 presence, evidenced by increased secretion of COL2 and COLX. Inhibition of TGFBR1 in JFLS-Ch results in overexpression of COL2. CONCLUSIONS: JFLS are chondrocyte-like, and Ch-conditioned media can abrogate this phenotype. The addition of exogenous BMP4 causes JFLS-Ch to restore this chondrocyte-like phenotype, suggesting that JFLS create a microenvironment favorable for endochondral bone formation, thereby contributing to joint growth disturbances in juvenile idiopathic arthritis.


Subject(s)
Bone Morphogenetic Protein 4 , Growth Disorders , Osteogenesis , Receptor, Transforming Growth Factor-beta Type I , Synoviocytes/metabolism , TGF-beta Superfamily Proteins/metabolism , Arthritis, Juvenile/complications , Arthritis, Juvenile/metabolism , Bone Morphogenetic Protein 4/antagonists & inhibitors , Bone Morphogenetic Protein 4/metabolism , Cells, Cultured , Cellular Microenvironment/drug effects , Cellular Microenvironment/physiology , Chondrocytes/physiology , Culture Media, Conditioned/pharmacology , Gene Expression Regulation , Growth Disorders/etiology , Growth Disorders/metabolism , Humans , Osteogenesis/drug effects , Osteogenesis/physiology , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/metabolism , Signal Transduction/drug effects
6.
Gene ; 787: 145627, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-33831497

ABSTRACT

The objective of this study was to identify the gonadal somatic cells in the Yesso scallop using a novel molecular marker. This study is the first to identify the bone morphogenetic protein 2a (Bmp2a) gene as a gonadal somatic cell-specific gene in this bivalve. We performed a transcriptomic survey to identify the transforming growth factor-ß (TGFß) superfamily members that act in Yesso scallop gonad development. BLAST survey, phylogenetic tree, and RT-PCR analyses screened BMP molecules (i.e., bmp2a and bmp10a), which are members of the TGFß superfamily that show gonad-specific expression. Among the BMPs from the Yesso scallop, in situ hybridization accompanied by RNAscope assay identified that bmp2a mRNA was specifically expressed in the gonadal somatic cells localized in the interspace between germ cells. Real-time quantitative PCR (qPCR) analysis revealed that bmp2a mRNA expression increased during the reproductive phase. The relative expression of bmp2a mRNA was lowest at the beginning of the growing stage and peaked at the mature stage in both sexes. These observations indicate that bmp2a-positive gonadal somatic cells support germ cell growth and differentiation during the reproductive phase for both sexes. This study provides new insights into gonadal somatic cell biology in marine invertebrates and we propose that TGFß signaling is necessary for gonad development in bivalves.


Subject(s)
Gonads/cytology , Gonads/metabolism , Pectinidae/metabolism , TGF-beta Superfamily Proteins/metabolism , Animals , Antigens, Differentiation , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Computer Simulation , Female , Genetic Markers , Gonads/growth & development , In Situ Hybridization , Male , Pectinidae/cytology , Pectinidae/genetics , Pectinidae/growth & development , Phylogeny , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reproduction , Signal Transduction , TGF-beta Superfamily Proteins/genetics , Tissue Distribution , Transcriptome
7.
Cell Signal ; 77: 109826, 2021 01.
Article in English | MEDLINE | ID: mdl-33160018

ABSTRACT

Most myocardial pathologic conditions are associated with cardiac fibrosis, the expansion of the cardiac interstitium through deposition of extracellular matrix (ECM) proteins. Although replacement fibrosis plays a reparative role after myocardial infarction, excessive, unrestrained or dysregulated myocardial ECM deposition is associated with ventricular dysfunction, dysrhythmias and adverse prognosis in patients with heart failure. The members of the Transforming Growth Factor (TGF)-ß superfamily are critical regulators of cardiac repair, remodeling and fibrosis. TGF-ßs are released and activated in injured tissues, bind to their receptors and transduce signals in part through activation of cascades involving a family of intracellular effectors the receptor-activated Smads (R-Smads). This review manuscript summarizes our knowledge on the role of Smad signaling cascades in cardiac fibrosis. Smad3, the best-characterized member of the family plays a critical role in activation of a myofibroblast phenotype, stimulation of ECM synthesis, integrin expression and secretion of proteases and anti-proteases. In vivo, fibroblast Smad3 signaling is critically involved in scar organization and exerts matrix-preserving actions. Although Smad2 also regulates fibroblast function in vitro, its in vivo role in rodent models of cardiac fibrosis seems more limited. Very limited information is available on the potential involvement of the Smad1/5/8 cascade in cardiac fibrosis. Dissection of the cellular actions of Smads in cardiac fibrosis, and identification of patient subsets with overactive or dysregulated myocardial Smad-dependent fibrogenic responses are critical for design of successful therapeutic strategies in patients with fibrosis-associated heart failure.


Subject(s)
Myocardial Infarction/pathology , Smad Proteins/metabolism , Animals , Diabetes Mellitus/pathology , Extracellular Matrix Proteins/metabolism , Humans , Myocardial Infarction/metabolism , Myofibroblasts/cytology , Myofibroblasts/metabolism , Signal Transduction , TGF-beta Superfamily Proteins/metabolism
8.
Genes (Basel) ; 11(3)2020 03 24.
Article in English | MEDLINE | ID: mdl-32213808

ABSTRACT

During embryonic development in vertebrates, morphogens play an important role in cell fate determination and morphogenesis. Bone morphogenetic proteins (BMPs) belonging to the transforming growth factor-ß (TGF-ß) family control the dorsal-ventral (DV) patterning of embryos, whereas other morphogens such as fibroblast growth factor (FGF), Wnt family members, and retinoic acid (RA) regulate the formation of the anterior-posterior (AP) axis. Activation of morphogen signaling results in changes in the expression of target genes including transcription factors that direct cell fate along the body axes. To ensure the correct establishment of the body plan, the processes of DV and AP axis formation must be linked and coordinately regulated by a fine-tuning of morphogen signaling. In this review, we focus on the interplay of various intracellular regulatory mechanisms and discuss how communication among morphogen signaling pathways modulates body axis formation in vertebrate embryos.


Subject(s)
Body Patterning , Cell Communication , TGF-beta Superfamily Proteins/metabolism , Wnt Proteins/metabolism , Animals , Gene Expression Regulation, Developmental , Humans , TGF-beta Superfamily Proteins/genetics , Wnt Proteins/genetics
9.
Reprod Fertil Dev ; 32(6): 539-552, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32024582

ABSTRACT

Survival of the embryo and establishment of a pregnancy is a critical period in the reproductive function of female cattle. This review examines how the transforming growth factor-ß (TGFB) superfamily (i.e. bone morphogenetic protein (BMP) 15, growth differentiation factor (GDF) 9, anti-Müllerian hormone (AMH)) and interferon-τ (IFNT) affect ovarian function and embryo development. The oocyte in a primary follicle secretes BMP15 and GDF9, which, together, organise the surrounding granulosa and theca cells into the oocyte-cumulus-follicle complex. At the same time, the granulosa secretes AMH, which affects the oocyte. This autocrine-paracrine dialogue between the oocyte and somatic cells continues throughout follicle development and is fundamental in establishing the fertilisation potential and embryo developmental competency of oocytes. The early bovine embryo secretes IFNT, which acts at the uterine endometrium, corpus luteum and blood leucocytes. IFNT is involved in the maternal recognition of pregnancy and immunomodulation to prevent rejection of the embryo, and supports progesterone secretion. Manipulation of BMP15, GDF9, AMH and IFNT in both invivo and invitro studies has confirmed their importance in reproductive function in female cattle. This review makes the case that a deeper understanding of the biology of BMP15, GDF9, AMH and IFNT will lead to new strategies to increase embryo survival and improve fertility in cattle. The enhancement of oocyte quality, early embryo development and implantation is considered necessary for the next step change in the efficiency of natural and assisted reproduction in cattle.


Subject(s)
Cell Communication , Embryonic Development , Fertility , Interferon Type I/metabolism , Ovary/metabolism , Pregnancy Proteins/metabolism , TGF-beta Superfamily Proteins/metabolism , Animals , Anti-Mullerian Hormone/metabolism , Bone Morphogenetic Protein 15/metabolism , Cattle , Female , Growth Differentiation Factor 9/metabolism , Pregnancy , Reproductive Techniques, Assisted/veterinary , Signal Transduction
10.
Genomics ; 112(1): 332-345, 2020 01.
Article in English | MEDLINE | ID: mdl-30779940

ABSTRACT

Guard hair and cashmere undercoat are developed from primary and secondary hair follicle, respectively. Little is known about the gene expression differences between primary and secondary hair follicle cycling. In this study, we obtained RNA-seq data from cashmere and milk goats grown at four different seasons. We studied the differentially expressed genes (DEGs) during the yearly hair follicle cycling, and between cashmere and milk goats. WNT, NOTCH, MAPK, BMP, TGFß and Hedgehog signaling pathways were involved in hair follicle cycling in both cashmere and milk goat. However, Milk goat DEGs between different months were significantly more than cashmere goat DEGs, with the largest difference being identified in December. Some expression dynamics were confirmed by quantitative PCR and western blot, and immunohistochemistry. This study offers new information sources related to hair follicle cycling in milk and cashmere goats, which could be applicable to improve the wool production and quality.


Subject(s)
Goats/genetics , Hair Follicle/metabolism , Transcriptome , Animals , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Forkhead Transcription Factors/metabolism , Genomics , Goats/metabolism , Hair Follicle/growth & development , RNA-Seq , Seasons , TGF-beta Superfamily Proteins/genetics , TGF-beta Superfamily Proteins/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
11.
Domest Anim Endocrinol ; 70: 106398, 2020 01.
Article in English | MEDLINE | ID: mdl-31677486

ABSTRACT

The aim of this study was to investigate some of the growth and transcriptional factors originating from oocytes and granulosa cells in follicular fluid and to identify the relationships between the basic blood metabolite-metabolic hormones and intrafollicular lipopolysaccharide (LPS) concentrations. Thirty cows included in the study were allocated into 2 groups comprising 15 cows with healthy preovulatory follicles (cyclic cows) and 15 cows with confirmed cystic follicles. The ovaries and uteri of all cows were assessed by transrectal ultrasonographic examination. Blood serum samples were collected at 15, 25, 35, 45, and 55 d after calving for analysis of nonesterified fatty acids, ß-hydroxybutyrate, insulin, glucose, IGF-I, ACTH, and cortisol. Ovaries and uteri were examined using transrectal ultrasound. Vaginal discharge was evaluated on the same days. Follicular fluid was also aspirated on days 35-55 from the healthy preovulatory follicles and cystic follicles using a transvaginal ovum pickup method. The densitometric levels of inhibin-α, growth and differentiation factor (GDF-9), bone morphogenetic protein (BMP-6), and GATA-4 and GATA-6 proteins were analyzed by the Western blotting technique; the concentrations of antimullerian hormone (AMH), IGF-I, estradiol-17 beta (E2), and progesterone (P4) were determined by ELISA; and the concentrations of LPS in the follicular fluid were measured by the Limulus amebocyte lysate test. The serum insulin, ACTH, and cortisol concentrations were higher in cystic cows than cyclic cows, but serum IGF-I concentrations were lower in cystic cows. The IGF-I concentrations of cystic follicular fluids were lower, whereas AMH levels were significantly greater than those of healthy preovulatory follicular fluids. The cystic follicles had significantly lower expression levels of GDF-9, BMP-6, GATA-4, and GATA-6; in contrast, inhibin-α expression and LPS concentrations were significantly higher than in healthy preovulatory follicles. The proportion of pathologic vaginal discharge within 25 d postpartum in cystic cows were higher than in the cyclic group. In conclusion, it is suggested that intrafollicular dysregulation of the transforming growth factor-ß superfamily, growth, and transcriptional factors is affected by high intrafollicular LPS concentrations and systemic metabolic changes and these disturbances may be responsible for the generation of ovarian cysts.


Subject(s)
Cattle Diseases/metabolism , Lipopolysaccharides/metabolism , Ovarian Cysts/veterinary , Ovarian Follicle/growth & development , TGF-beta Superfamily Proteins/metabolism , Transcription Factors/metabolism , Animals , Blood Glucose , Cattle , Cattle Diseases/blood , Female , Gene Expression Regulation , Ovarian Cysts/metabolism , TGF-beta Superfamily Proteins/genetics , Transcription Factors/genetics
12.
Mol Cell ; 76(5): 753-766.e6, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31563432

ABSTRACT

The gene expression programs that define the identity of each cell are controlled by master transcription factors (TFs) that bind cell-type-specific enhancers, as well as signaling factors, which bring extracellular stimuli to these enhancers. Recent studies have revealed that master TFs form phase-separated condensates with the Mediator coactivator at super-enhancers. Here, we present evidence that signaling factors for the WNT, TGF-ß, and JAK/STAT pathways use their intrinsically disordered regions (IDRs) to enter and concentrate in Mediator condensates at super-enhancers. We show that the WNT coactivator ß-catenin interacts both with components of condensates and DNA-binding factors to selectively occupy super-enhancer-associated genes. We propose that the cell-type specificity of the response to signaling is mediated in part by the IDRs of the signaling factors, which cause these factors to partition into condensates established by the master TFs and Mediator at genes with prominent roles in cell identity.


Subject(s)
Enhancer Elements, Genetic/genetics , Mediator Complex/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Gene Expression Regulation/physiology , Humans , Intrinsically Disordered Proteins/metabolism , Mediator Complex/physiology , STAT Transcription Factors/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Smad3 Protein/metabolism , TGF-beta Superfamily Proteins/metabolism , Transcription, Genetic , Wnt Signaling Pathway , beta Catenin/metabolism
13.
Biochim Biophys Acta Mol Basis Dis ; 1864(9 Pt B): 3022-3037, 2018 09.
Article in English | MEDLINE | ID: mdl-29928977

ABSTRACT

The pathomechanisms underlying oxidative phosphorylation (OXPHOS) diseases are not well-understood, but they involve maladaptive changes in mitochondria-nucleus communication. Many studies on the mitochondria-nucleus cross-talk triggered by mitochondrial dysfunction have focused on the role played by regulatory proteins, while the participation of miRNAs remains poorly explored. MELAS (mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes) is mostly caused by mutation m.3243A>G in mitochondrial tRNALeu(UUR) gene. Adverse cardiac and neurological events are the commonest causes of early death in m.3243A>G patients. Notably, the incidence of major clinical features associated with this mutation has been correlated to the level of m.3243A>G mutant mitochondrial DNA (heteroplasmy) in skeletal muscle. In this work, we used a transmitochondrial cybrid model of MELAS (100% m.3243A>G mutant mitochondrial DNA) to investigate the participation of miRNAs in the mitochondria-nucleus cross-talk associated with OXPHOS dysfunction. High-throughput analysis of small-RNA-Seq data indicated that expression of 246 miRNAs was significantly altered in MELAS cybrids. Validation of selected miRNAs, including miR-4775 and miR-218-5p, in patient muscle samples revealed miRNAs whose expression declined with high levels of mutant heteroplasmy. We show that miR-218-5p and miR-4775 are direct regulators of fetal cardiac genes such as NODAL, RHOA, ISL1 and RXRB, which are up-regulated in MELAS cybrids and in patient muscle samples with heteroplasmy above 60%. Our data clearly indicate that TGF-ß superfamily signaling and an epithelial-mesenchymal transition-like program are activated in MELAS cybrids, and suggest that down-regulation of miRNAs regulating fetal cardiac genes is a risk marker of heart failure in patients with OXPHOS diseases.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Heart Failure/genetics , MELAS Syndrome/genetics , MicroRNAs/genetics , Myocardium/pathology , RNA, Transfer, Leu/genetics , Cell Line, Tumor , DNA, Mitochondrial/genetics , Datasets as Topic , Down-Regulation , Gene Expression Regulation, Developmental , Heart/growth & development , Heart Failure/pathology , High-Throughput Nucleotide Sequencing , Humans , MELAS Syndrome/complications , MELAS Syndrome/pathology , MicroRNAs/metabolism , Mitochondria/genetics , Mitochondria/pathology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mutation , Myocardium/cytology , Myocardium/metabolism , Oxidative Phosphorylation , Sequence Analysis, RNA , Signal Transduction/genetics , TGF-beta Superfamily Proteins/genetics , TGF-beta Superfamily Proteins/metabolism , Up-Regulation
14.
Cell Mol Biol (Noisy-le-grand) ; 64(15): 1-6, 2018 Dec 31.
Article in English | MEDLINE | ID: mdl-30672446

ABSTRACT

Based on the exciting insights gleaned from decades of ground-breaking research, it has become evident that deregulated signaling pathways play instrumental role in cancer development and progression. Interestingly discovery of non-coding RNAs has revolutionized our understanding related to transcription, post-transcription and translation. Modern era has witnessed landmark discoveries in the field of molecular cancer and non-coding RNA biology has undergone tremendous broadening. There has been an exponential growth in the list of publications related to non-coding RNAs and overwhelmingly increasing classes of non-coding RNAs are adding new layers of complexity to already complicated nature of cancer. Regulation of TGF/SMAD signaling by miRNAs and LncRNAs has opened new horizons for therapeutic targeting of TGF/SMAD pathway. In this review we have set spotlight on central role of LncRNAs in modulation of TGF/SMAD pathway. Major proportion of the available evidence is underlining positive role of LncRNAs in contextual regulation of TGF/SMAD pathway. LncRNAs are vital to these regulatory networks because they provide a background support to make the TGF/SMAD mediated intracellular signaling more smooth or make transduction cascade more flexible in response to cues from extracellular environment. Therefore, in accordance with this notion, MALAT1, OIP5-AS1, MIR100HG, HOTAIR, ANRIL, PVT1, AFAP1-AS1, SPRY4-IT, ZEB2NAT, TUG1 and Lnc-SNHG1 have been reported to positively regulate TGF/SMAD signaling. In this review, we have focused on the regulation of TGF/SMAD signaling by LncRNAs and how these non-coding RNAs can be therapeutically exploited. Short-interfering RNA (siRNA) and natural products are currently being tested for efficacy against different LncRNAs. Nanotechnological strategies to efficiently deliver LncRNA-targeting siRNAs are also currently being investigated in different cancers.


Subject(s)
Neoplasms/genetics , Neoplasms/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction , Smad Proteins/metabolism , TGF-beta Superfamily Proteins/metabolism , Animals , Humans , Models, Biological
15.
Biomaterials ; 104: 279-96, 2016 10.
Article in English | MEDLINE | ID: mdl-27474964

ABSTRACT

The momentum to compose this Leading Opinion on the synergistic induction of bone formation suddenly arose when a simple question was formulated during a discussion session on how to boost the often limited induction of bone formation seen in clinical contexts. Re-examination of morphological and molecular data available on the rapid induction of bone formation by the recombinant human transforming growth factor-ß3 (hTGF-ß3) shows that hTGF-ß3 replicates the synergistic induction of bone formation as invocated by binary applications of hOP-1:hTGF-ß1 at 20:1 by weight when implanted in heterotopic sites of the rectus abdominis muscle of the Chacma baboon, Papio ursinus. The rapid induction of bone formation in primates by hTGF-ß3 may stem from bursts of cladistic evolution, now redundant in lower animal species but still activated in primates by relatively high doses of hTGF-ß3. Contrary to rodents, lagomorphs and canines, the three mammalian TGF-ß isoforms induce rapid and substantial bone formation when implanted in heterotopic rectus abdominis muscle sites of P. ursinus, with unprecedented regeneration of full thickness mandibular defects with rapid mineralization and corticalization. Provocatively, thus providing potential molecular and biological rationales for the apparent redundancy of osteogenic molecular signals in primates, binary applications of recombinant human osteogenic protein-1 (hOP-1) with low doses of hTGF-ß1 and -ß3, synergize to induce massive ossicles in heterotopic rectus abdominis, orthotopic calvarial and mandibular sites of P. ursinus. The synergistic binary application of homologous but molecularly different soluble molecular signals has indicated that per force several secreted molecular signals are required singly, synchronously and synergistically to induce optimal osteogenesis. The morphological hallmark of the synergistic induction of bone formation is the rapid differentiation of large osteoid seams enveloping haematopoietic bone marrow that forms by day 15 in heterotopic rectus abdominis sites. Synergistic binary applications also induce the morphogenesis of rudimentary embryonic growth plates indicating that the "memory" of developmental events in embryo can be redeployed postnatally by the application of morphogen combinations. Synergistic binary applications or single relatively high doses of hTGF-ß3 have shown that hTGF-ß3 induces bone by expressing a variety of inductive morphogenetic proteins that result in the rapid induction of bone formation. Tissue induction thus invocated singly by hTGF-ß3 recapitulates the synergistic induction of bone formation by binary applications of hTGF-ß1 and -ß3 isoforms with hOP-1. Both synergistic strategies result in the rapid induction and expansion of the transformed mesenchymal tissue into large corticalized heterotopic ossicles with osteoblast-like cell differentiation at the periphery of the implanted reconstituted specimens with "tissue transfiguration" in vivo. Molecularly, the rapid induction of bone formation by binary applications of hOP-1 and hTGF-ß3 or by hTGF-ß3 applied singly resides in the up-regulation of selected genes involved in tissue induction and morphogenesis, Osteocalcin, RUNX-2, OP-1, TGF-ß1 and -ß3 with however the noted lack of TGF-ß2 up-regulation.


Subject(s)
Bone Development/physiology , Bone Regeneration/physiology , Osteogenesis/physiology , TGF-beta Superfamily Proteins/metabolism , Animals , Dogs , Humans , Multigene Family/physiology , Species Specificity
16.
Neural Dev ; 11: 9, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27048518

ABSTRACT

BACKGROUND: Brain size and patterning are dependent on dosage-sensitive morphogen signaling pathways - yet how these pathways are calibrated remains enigmatic. Recent studies point to a new role for microRNAs in tempering the spatio-temporal range of morphogen functions during development. Here, we investigated the role of miR-135a, derived from the mir-135a-2 locus, in embryonic forebrain development. METHOD: 1. We characterized the expression of miR-135a, and its host gene Rmst, by in situ hybridization (ish). 2. We conditionally ablated, or activated, beta-catenin in the dorsal forebrain to determine if this pathway was necessary and/or sufficient for Rmst/miR-135a expression. 3. We performed bioinformatics analysis to unveil the most predicted pathways targeted by miR-135a. 4. We performed gain and loss of function experiments on mir-135a-2 and analyzed by ish the expression of key markers of cortical hem, choroid plexus, neocortex and hippocampus. RESULTS: 1. miR-135a, embedded in the host long non-coding transcript Rmst, is robustly expressed, and functional, in the medial wall of the embryonic dorsal forebrain, a Wnt and TGFß/BMP-rich domain. 2. Canonical Wnt/beta-catenin signaling is critical for the expression of Rmst and miR-135a, and the cortical hem determinant Lmx1a. 3. Bioinformatics analyses reveal that the Wnt and TGFß/BMP cascades are among the top predicted pathways targeted by miR-135a. 4. Analysis of mir-135a-2 null embryos showed that dorsal forebrain development appeared normal. In contrast, modest mir-135a-2 overexpression, in the early dorsal forebrain, resulted in a phenotype resembling that of mutants with Wnt and TGFß/BMP deficits - a smaller cortical hem and hippocampus primordium associated with a shorter neocortex as well as a less convoluted choroid plexus. Interestingly, late overexpression of mir-135a-2 revealed no change. CONCLUSIONS: All together, our data suggests the existence of a Wnt/miR-135a auto-regulatory loop, which could serve to limit the extent, the duration and/or intensity of the Wnt and, possibly, the TGFß/BMP pathways.


Subject(s)
MicroRNAs/metabolism , Prosencephalon/embryology , Prosencephalon/metabolism , Wnt Signaling Pathway , Animals , LIM-Homeodomain Proteins/metabolism , Mice , TGF-beta Superfamily Proteins/metabolism , Transcription Factors/metabolism
17.
PLoS One ; 10(12): e0145322, 2015.
Article in English | MEDLINE | ID: mdl-26683658

ABSTRACT

Genomic technologies including microarrays and next-generation sequencing have enabled the generation of molecular signatures of prostate cancer. Lists of differentially expressed genes between malignant and non-malignant states are thought to be fertile sources of putative prostate cancer biomarkers. However such lists of differentially expressed genes can be highly variable for multiple reasons. As such, looking at differential expression in the context of gene sets and pathways has been more robust. Using next-generation genome sequencing data from The Cancer Genome Atlas, differential gene expression between age- and stage- matched human prostate tumors and non-malignant samples was assessed and used to craft a pathway signature of prostate cancer. Up- and down-regulated genes were assigned to pathways composed of curated groups of related genes from multiple databases. The significance of these pathways was then evaluated according to the number of differentially expressed genes found in the pathway and their position within the pathway using Gene Set Enrichment Analysis and Signaling Pathway Impact Analysis. The "transforming growth factor-beta signaling" and "Ran regulation of mitotic spindle formation" pathways were strongly associated with prostate cancer. Several other significant pathways confirm reported findings from microarray data that suggest actin cytoskeleton regulation, cell cycle, mitogen-activated protein kinase signaling, and calcium signaling are also altered in prostate cancer. Thus we have demonstrated feasibility of pathway analysis and identified an underexplored area (Ran) for investigation in prostate cancer pathogenesis.


Subject(s)
Prostatic Neoplasms/metabolism , Aged , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Ontology , Humans , Male , Middle Aged , Prostatic Neoplasms/genetics , Signal Transduction , TGF-beta Superfamily Proteins/genetics , TGF-beta Superfamily Proteins/metabolism , Transcriptome
18.
Sci Transl Med ; 7(299): 299rv4, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26246170

ABSTRACT

Immunological and inflammatory processes downstream of dystrophin deficiency as well as metabolic abnormalities, defective autophagy, and loss of regenerative capacity all contribute to muscle pathology in Duchenne muscular dystrophy (DMD). These downstream cascades offer potential avenues for pharmacological intervention. Modulating the inflammatory response and inducing immunological tolerance to de novo dystrophin expression will be critical to the success of dystrophin-replacement therapies. This Review focuses on the role of the inflammatory response in DMD pathogenesis and opportunities for clinical intervention.


Subject(s)
Immunity, Innate , Muscular Dystrophy, Duchenne , Cytokines/metabolism , Dystrophin/metabolism , Fibrosis , Humans , Inflammation , Muscular Dystrophy, Duchenne/etiology , Muscular Dystrophy, Duchenne/immunology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Signal Transduction , TGF-beta Superfamily Proteins/metabolism
19.
Genes Chromosomes Cancer ; 54(7): 453-62, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25931269

ABSTRACT

As a transforming growth factor-ß (TGF-ß)-inducible gene, the expression of Krüppel-like transcription factor 11 (KLF11) is altered in several types of cancer. In the current study, through using human 9K CpG island array, KLF11 was identified as one of hypermethylated genes in RAS-transformed ovarian T29H cells. Methylation of the KLF11 promoter was also observed in ovarian cancer tissue samples accompanied by significantly reduced KLF11 gene expression. Interestingly, the expression of SMAD2, SMAD3, and SMAD7 genes was reduced in the tumour, whilst no change was found in TGF-ß expression. Our data suggest a relationship between promoter DNA methylation and KLF11 gene expression in ovarian cancer tumorigenesis.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Methylation , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism , Aged , Apoptosis Regulatory Proteins , Carcinoma, Ovarian Epithelial , Cell Cycle Proteins/genetics , Cell Line, Tumor , China , CpG Islands , Female , Genetic Association Studies , Humans , Middle Aged , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Repressor Proteins/genetics , Smad Proteins/metabolism , TGF-beta Superfamily Proteins/metabolism
20.
Horm Mol Biol Clin Investig ; 18(2): 53-61, 2014 May.
Article in English | MEDLINE | ID: mdl-25390002

ABSTRACT

Aldosterone regulates sodium (Na+) and potassium (K+) transports in epithelial cells. Besides, aldosterone participates in cardiac alterations associated with hypertension, heart failure, diabetes, and other pathological alterations. One of the main cardiac alterations induced by aldosterone is cardiac hypertrophy in which different mechanisms are involved such as increased cardiomyocyte, calcium concentration, oxidative stress, and inflammatory and fibrotic mediators stimulation. Many epidemiological studies have demonstrated that left ventricular hypertrophy is associated with significantly increased risk of heart failure and malignant arrhythmias. SGK1 is a member of the serine/threonine kinase gene family that plays an important role in the absorption of Na+ and water through the Na+ channel in the apical membrane of tubular epithelial cells. SGK1 has been related to fibrotic mediator increase such as connective tissue growth factor (CTGF) and transforming growth factor-ß (TGF-ß) as well as inflammatory [tumor necrosis factor-α (TNF-α) and interleukin (IL)-1ß] and oxidative (NADPH oxidase) species. It has been shown that aldosterone induces SGK1 gene expression not only in kidneys but also in the heart. Supporting the central role of SGK1 in cardiac alterations induced by aldosterone, treatment with the mineralocorticoid antagonist spironolactone is able to reduce the gene expression of SGK1 in aldosterone-treated rats. Taken together, data suggest the involvement of SGK1 in a complex intracellular signaling, involving fibrotic, inflammatory, and oxidative pathways, which lead to cardiac hypertrophy and fibrosis induced by aldosterone.


Subject(s)
Aldosterone/metabolism , Immediate-Early Proteins/metabolism , Myocardium/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Connective Tissue Growth Factor/metabolism , Endothelium, Vascular/metabolism , Fibrosis , Gene Expression , Humans , Inflammation/metabolism , Inflammation/pathology , Myocardium/pathology , NADPH Oxidases/metabolism , Oxidative Stress , Signal Transduction , TGF-beta Superfamily Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...