3.Spectrum of Novel Hereditary Hemorrhagic Telangiectasia Variants in an Austrian Patient Cohort
Martin KOENIGHOFER ; Thomas PARZEFALL ; Alexandra FROHNE ; Matthew ALLEN ; Ursula UNTERBERGER ; Franco LACCONE ; Christian SCHOEFER ; Klemens FREI ; Trevor LUCAS
Clinical and Experimental Otorhinolaryngology 2019;12(4):405-411
OBJECTIVES: Hereditary hemorrhagic telangiectasia (HHT) is a rare autosomal dominant genetic disorder characterized by pathogenic blood vessel development and maintenance. HHT type 1 (HHT1) and type 2 (HHT2) are caused by variants in endoglin (ENG) and activin receptor-like kinase-1 (ACVRL1), respectively. The aim of this study was to identify the spectrum of pathogenic variants in ENG and ACVRL1 in Austrian HHT families. METHODS: In this prospective study, eight Austrian HHT families were screened for variants in ENG and ACVRL1 by polymerase chain reaction amplification and sequencing of DNA isolated from peripheral blood. RESULTS: Heterozygous variants were identified in all families under study. HHT1 was caused by a novel c.816+1G>A splice donor variant, a novel c.1479C>A nonsense (p.Cys493X) variant and a published c.1306C>T nonsense (p.Gln436X) variant in ENG. Variants found in ACVRL1 were novel c.200G>C (p.Arg67Pro) and known c.772G>A (p.Gly258Ser) missense variants in highly conserved residues, a known heterozygous c.100dupT frameshift (p.Cys34Leufs*4) and the known c.1204G>A missense (p.Gly402Ser) and c.1435C>T nonsense (p.Arg479X) variants as causes of HHT2. CONCLUSION: Novel and published variants in ENG (37.5%) and ACVRL1 (62.5%) were exclusively identified as the cause of HHT in an Austrian patient cohort. Identification of novel causative genetics variants should facilitate the development of tailored therapeutical applications in the future treatment of autosomal dominant HHT.
Activins
;
Blood Vessels
;
Cohort Studies
;
DNA
;
Genetics
;
Humans
;
Polymerase Chain Reaction
;
Prospective Studies
;
Telangiectasia, Hereditary Hemorrhagic
;
Telangiectasis
;
Tissue Donors
4.Enhanced Anti-Cancer Effects of Conditioned Medium from Hypoxic Human Umbilical Cord–Derived Mesenchymal Stem Cells
Kyu Hyun HAN ; Ae Kyeong KIM ; Gun Jae JEONG ; Hye Ran JEON ; Suk Ho BHANG ; Dong Ik KIM
International Journal of Stem Cells 2019;12(2):291-303
BACKGROUND AND OBJECTIVES: There have been contradictory reports on the pro-cancer or anti-cancer effects of mesenchymal stem cells. In this study, we investigated whether conditioned medium (CM) from hypoxic human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) (H-CM) showed enhanced anti-cancer effects compared with CM from normoxic hUC-MSCs (N-CM). METHODS AND RESULTS: Compared with N-CM, H-CM not only strongly reduced cell viability and increased apoptosis of human cervical cancer cells (HeLa cells), but also increased caspase-3/7 activity, decreased mitochondrial membrane potential (MMP), and induced cell cycle arrest. In contrast, cell viability, apoptosis, MMP, and cell cycle of human dermal fibroblast (hDFs) were not significantly changed by either CM whereas caspase-3/7 activity was decreased by H-CM. Protein antibody array showed that activin A, Beta IG-H3, TIMP-2, RET, and IGFBP-3 were upregulated in H-CM compared with N-CM. Intracellular proteins that were upregulated by H-CM in HeLa cells were represented by apoptosis and cell cycle arrest terms of biological processes of Gene Ontology (GO), and by cell cycle of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. In hDFs, negative regulation of apoptosis in biological process of GO and PI3K-Akt signaling pathway of KEGG pathways were represented. CONCLUSIONS: H-CM showed enhanced anti-cancer effects on HeLa cells but did not influence cell viability or apoptosis of hDFs and these different effects were supported by profiling of secretory proteins in both kinds of CM and intracellular signaling of HeLa cells and hDFs.
Activins
;
Anoxia
;
Apoptosis
;
Biological Processes
;
Cell Cycle
;
Cell Cycle Checkpoints
;
Cell Survival
;
Culture Media, Conditioned
;
Fibroblasts
;
Gene Ontology
;
Genome
;
HeLa Cells
;
Humans
;
Insulin-Like Growth Factor Binding Protein 3
;
Membrane Potential, Mitochondrial
;
Mesenchymal Stromal Cells
;
Tissue Inhibitor of Metalloproteinase-2
;
Uterine Cervical Neoplasms
5.TGF-β induces Smad2 Phosphorylation, ARE Induction, and Trophoblast Differentiation
Renee E ALBERS ; Kaisa SELESNIEMI ; David R C NATALE ; Thomas L BROWN
International Journal of Stem Cells 2018;11(1):111-120
BACKGROUND: Transforming growth factor beta (TGF-β) signaling has been shown to control a large number of critical cellular actions such as cell death, differentiation, and development and has been implicated as a major regulator of placental function. SM10 cells are a mouse placental progenitor cell line, which has been previously shown to differentiate into nutrient transporting, labyrinthine-like cells upon treatment with TGF-β. However, the signal transduction pathway activated by TGF-β to induce SM10 progenitor differentiation has yet to be fully investigated. MATERIALS AND METHODS: In this study the SM10 labyrinthine progenitor cell line was used to investigate TGF-β induced differentiation. Activation of the TGF-β pathway and the ability of TGF-β to induce differentiation were investigated by light microscopy, luciferase assays, and Western blot analysis. RESULTS AND CONCLUSIONS: In this report, we show that three isoforms of TGF-β have the ability to terminally differentiate SM10 cells, whereas other predominant members of the TGF-β superfamily, Nodal and Activin A, do not. Additionally, we have determined that TGF-β induced Smad2 phosphorylation can be mediated via the ALK-5 receptor with subsequent transactivation of the Activin response element. Our studies identify an important regulatory signaling pathway in SM10 progenitor cells that is involved in labyrinthine trophoblast differentiation.
Activins
;
Animals
;
Blotting, Western
;
Cell Death
;
Luciferases
;
Mice
;
Microscopy
;
Phosphorylation
;
Placenta
;
Protein Isoforms
;
Response Elements
;
Signal Transduction
;
Stem Cells
;
Transcriptional Activation
;
Transforming Growth Factor beta
;
Trophoblasts
6.Pluripotent stem cells secrete Activin A to improve their epiblast competency after injection into recipient embryos.
Jinzhu XIANG ; Suying CAO ; Liang ZHONG ; Hanning WANG ; Yangli PEI ; Qingqing WEI ; Bingqiang WEN ; Haiyuan MU ; Shaopeng ZHANG ; Liang YUE ; Genhua YUE ; Bing LIM ; Jianyong HAN
Protein & Cell 2018;9(8):717-728
It is not fully clear why there is a higher contribution of pluripotent stem cells (PSCs) to the chimera produced by injection of PSCs into 4-cell or 8-cell stage embryos compared with blastocyst injection. Here, we show that not only embryonic stem cells (ESCs) but also induced pluripotent stem cells (iPSCs) can generate F0 nearly 100% donor cell-derived mice by 4-cell stage embryo injection, and the approach has a "dose effect". Through an analysis of the PSC-secreted proteins, Activin A was found to impede epiblast (EPI) lineage development while promoting trophectoderm (TE) differentiation, resulting in replacement of the EPI lineage of host embryos with PSCs. Interestingly, the injection of ESCs into blastocysts cultured with Activin A (cultured from 4-cell stage to early blastocyst at E3.5) could increase the contribution of ESCs to the chimera. The results indicated that PSCs secrete protein Activin A to improve their EPI competency after injection into recipient embryos through influencing the development of mouse early embryos. This result is useful for optimizing the chimera production system and for a deep understanding of PSCs effects on early embryo development.
Activins
;
metabolism
;
Animals
;
Cells, Cultured
;
Embryonic Development
;
Germ Layers
;
metabolism
;
Mice
;
Pluripotent Stem Cells
;
cytology
;
metabolism
7.In vitro differentiation of spermatogonial stem cells using testicular cells from Guangxi Bama mini-pig
Huimin ZHAO ; Junyu NIE ; Xiangxing ZHU ; Yangqing LU ; Xingwei LIANG ; Huiyan XU ; Xiaogan YANG ; Yunkai ZHANG ; Kehuan LU ; Shengsheng LU
Journal of Veterinary Science 2018;19(5):592-599
In this study, we attempted to establish a culture system for in vitro spermatogenesis from spermatogonial stem cells (SSCs) of Bama mini-pig. Dissociated testicular cells from 1-month-old pigs were co-cultured to mimic in vivo spermatogenesis. The testicular cells were seeded in minimum essential medium alpha (α-MEM) supplemented with Knockout serum replacement (KSR). Three-dimensional colonies formed after 10 days of culture. The colonies showed positive staining for SSC-associated markers such as UCHL1, PLZF, THY1, OCT4, Dolichos biflorus agglutinin, and alkaline phosphatase. Induction of SSCs was performed in α-MEM + KSR supplemented with retinoic acid, bone morphogenetic protein 4, activin A, follicle-stimulating hormone, or testosterone. The results showed that STRA8, DMC1, PRM1, and TNP1 were upregulated significantly in the colonies after induction compared to that in testis from 1-month-old pigs, while expression levels of those genes were significantly low compared to those in 2-month-old testis. However, upregulation of ACROSIN was not significant. Replacement of α-MEM and KSR with Iscove's modified Dulbecco's medium and fetal bovine serum did not upregulate expression of these genes significantly. These results indicate that SSCs of Bama mini-pig could undergo differentiation and develop to a post-meiotic stage in α-MEM supplemented with KSR and induction factors.
Acrosin
;
Activins
;
Alkaline Phosphatase
;
Bone Morphogenetic Protein 4
;
Dolichos
;
Follicle Stimulating Hormone
;
Humans
;
In Vitro Techniques
;
Infant
;
Infant, Newborn
;
Spermatogenesis
;
Stem Cells
;
Swine
;
Testis
;
Testosterone
;
Tretinoin
;
Up-Regulation
8.Recent Topics in Fibrodysplasia Ossificans Progressiva.
Takenobu KATAGIRI ; Sho TSUKAMOTO ; Yutaka NAKACHI ; Mai KURATANI
Endocrinology and Metabolism 2018;33(3):331-338
Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disease that is characterized by the formation of heterotopic bone tissues in soft tissues, such as skeletal muscle, ligament, and tendon. It is difficult to remove such heterotopic bones via internal medicine or invasive procedures. The identification of activin A receptor, type I (ACVR1)/ALK2 gene mutations associated with FOP has allowed the genetic diagnosis of FOP. The ACVR1/ALK2 gene encodes the ALK2 protein, which is a transmembrane kinase receptor in the transforming growth factor-β family. The relevant mutations activate intracellular signaling in vitro and induce heterotopic bone formation in vivo. Activin A is a potential ligand that activates mutant ALK2 but not wild-type ALK2. Various types of small chemical and biological inhibitors of ALK2 signaling have been developed to establish treatments for FOP. Some of these are in clinical trials in patients with FOP.
Activins
;
Bone and Bones
;
Diagnosis
;
Humans
;
In Vitro Techniques
;
Internal Medicine
;
Ligaments
;
Muscle, Skeletal
;
Myositis Ossificans*
;
Osteogenesis
;
Phosphotransferases
;
Tendons
;
Transforming Growth Factor beta
9.Interactive Roles of Activin A in Epidermal Regeneration.
Jee Woong CHOI ; Kyung Mi NAM ; Hye Ryung CHOI ; Chang Hun HUH ; Kyung Chan PARK
Annals of Dermatology 2018;30(6):755-757
No abstract available.
Activins*
;
Regeneration*
10.Activin A/BMP2 Chimera (AB204) Exhibits Better Spinal Bone Fusion Properties than rhBMP2
Dalsung RYU ; Byung Hak YOON ; Chang Hyun OH ; Moon Hang KIM ; Ji Yong KIM ; Seung Hwan YOON ; Senyon CHOE
Journal of Korean Neurosurgical Society 2018;61(6):669-679
OBJECTIVE: To compare the spinal bone fusion properties of activin A/BMP2 chimera (AB204) with recombinant human bone morphogenetic protein (rhBMP2) using a rat posterolateral spinal fusion model.METHODS: The study was designed to compare the effects and property at different dosages of AB204 and rhBMP2 on spinal bone fusion. Sixty-one male Sprague-Dawley rats underwent posterolateral lumbar spinal fusion using one of nine treatments during the study, that is, sham; osteon only; 3.0 μg, 6.0 μg, or 10.0 μg of rhBMP2 with osteon; and 1.0 μg, 3.0 μg, 6.0 μg, or 10.0 μg of AB204 with osteon. The effects and property on spinal bone fusion was calculated at 4 and 8 weeks after treatment using the scores of physical palpation, simple radiograph, micro-computed tomography, and immunohistochemistry.RESULTS: Bone fusion scores were significantly higher for 10.0 μg AB204 and 10.0 μg rhBMP2 than for osteon only or 1.0 μg AB204. AB204 exhibited more prolonged osteoblastic activity than rhBMP2. Bone fusion properties of AB204 were similar with the properties of rhBMP2 at doses of 6.0 and 10.0 μg, but, the properties of AB204 at doses of 3.0 μg exhibited better than the properties of rhBMP2 at doses of 3.0 μg.CONCLUSION: AB204 chimeras could to be more potent for treating spinal bone fusion than rhBMP2 substitutes with increased osteoblastic activity for over a longer period.
Activins
;
Animals
;
Bone Morphogenetic Proteins
;
Chimera
;
Haversian System
;
Humans
;
Immunohistochemistry
;
Male
;
Osteoblasts
;
Palpation
;
Rats
;
Rats, Sprague-Dawley
;
Spinal Fusion

Result Analysis
Print
Save
E-mail