1
|
Lou H, Lu H, Zhang S, Shi Y, Xu E, Liu D, Chen Q. Highly aligned myotubes formation of piscine satellite cells in 3D fibrin hydrogels of cultured meat. Int J Biol Macromol 2024; 282:136879. [PMID: 39490877 DOI: 10.1016/j.ijbiomac.2024.136879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Currently, various cultured meat products, including chicken, beef and pork, have been developed. However, established methods for production cultured fish meat with highly aligned myotubes are still lack. In this study, we introduced a culture method based on high-biocompatibility fibrin hydrogels with an easy-to-use tissue mold for obtaining cultured fish fillets that closely mimicked the structure of natural fish fillets. Results showed that highly aligned myotubes were observed within the muscle bundles culturing in the tissue mold. The myotube fusion index was also increased to 72.65 %. Furthermore, key differentiation genes (desmin, myosin light chain kinase, myocilin) were up-regulated in the tissue mold group. Transcriptomic analysis further supported the effectiveness of method in promoting myoblast fusion. Stiffness of the muscle bundles was also positively impacted by the tissue mold. Ultimately, sensory and nutritional characteristics of natural and cultured fish fillets were compared, revealing that cultured fish fillets prepared from the tissue mold was closer to natural fish fillets in sensory characteristic, and there were still some gaps with natural fish fillets in nutritional characteristic. Overall, our findings suggest that optimizing culture methods can help bridge some gaps between natural meat and cultured meat, facilitating the development of cultured fish meat.
Collapse
Affiliation(s)
- Hanghang Lou
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Hongyun Lu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Shengliang Zhang
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Ying Shi
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
| | - Enbo Xu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China; Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Donghong Liu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China; Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Qihe Chen
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China; Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| |
Collapse
|
2
|
Onikanni SA, Yang CY, Noriega L, Wang CH. U0126 Compound Triggers Thermogenic Differentiation in Preadipocytes via ERK-AMPK Signaling Axis. Int J Mol Sci 2023; 24:ijms24097987. [PMID: 37175694 PMCID: PMC10178890 DOI: 10.3390/ijms24097987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
In recent years, thermogenic differentiation and activation in brown and white adipose tissues have been regarded as one of the major innovative and promising strategies for the treatment and amelioration of obesity. However, the pharmacological approach towards this process has had limited and insufficient commitments, which presents a greater challenge for obesity treatment. This research evaluates the effects of U0126 compound on the activation of thermogenic differentiation during adipogenesis. The results show that U0126 pretreatment primes both white and brown preadipocytes to upregulate thermogenic and mitochondrial genes as well as enhance functions during the differentiation process. We establish that U0126-mediated thermogenic differentiation induction occurs partially via AMPK activation signaling. The findings of this research suggest U0126 as a promising alternative ligand in pursuit of a pharmacological option to increase thermogenic adipocyte formation and improve energy expenditure. Thus it could pave the way for the discovery of therapeutic drugs for the treatment of obesity and its related complications.
Collapse
Affiliation(s)
- Sunday Amos Onikanni
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
- Department of Chemical Sciences, Biochemistry Unit, Afe Babalola University, Ado-Ekiti 360101, Ekiti State, Nigeria
| | - Cheng-Ying Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
| | - Lloyd Noriega
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
| | - Chih-Hao Wang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 406040, Taiwan
- Graduate Institute of Cell Biology, College of Life Sciences, China Medical University, Taichung 406040, Taiwan
| |
Collapse
|
3
|
Di Rocco A, Camero S, Benedetti A, Lozanoska-Ochser B, Megiorni F, Marchese C, Stramucci L, Ciccarelli C, Bouché M, Bossi G, Marampon F, Zani BM. Anti‑oncogenic and pro‑myogenic action of the MKK6/p38/AKT axis induced by targeting MEK/ERK in embryonal rhabdomyosarcoma. Oncol Rep 2022; 48:151. [PMID: 35801577 PMCID: PMC9350981 DOI: 10.3892/or.2022.8363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/16/2022] [Indexed: 11/05/2022] Open
Abstract
Insights into the molecular and cellular biology of embryonal rhabdomyosarcoma (ERMS), an aggressive paediatric tumour, are required in order to identify new targets for novel treatments that may benefit patients with this disease. The present study examined the functional effects of MKK3 and MKK6, two upstream kinases of p38, and found that the ectopic expression of MKK6 led to rapid p38 activation and the myogenic differentiation of ERMS cells, whereas MKK3 failed to induce differentiation, while maintaining the proliferation state. Myogenin and myosin heavy chain were induced in MKK6‑overexpressing ERMS cells and were inhibited by the p38 inhibitor, SB203580. The expression of Myc and ERK‑PO4 increased under the effect of SB203580, whereas it decreased in MKK6‑overexpressing cells. AKT activation was part of the myogenic program triggered by MKK6 overexpression alone. To the best of our knowledge, the present study demonstrates, for the first time, that the endogenous MKK6 pathway may be recovered by MEK/ERK inhibition (U0126 and trametinib) and that it concomitantly induces the reversal of the oncogenic pattern and the induction of the myogenic differentiation of ERMS cell lines. The effects of MEK/ERK inhibitors markedly increase the potential clinical applications in ERMS, particularly on account of the MEK inhibitor‑induced early MKK6/p38 axis activation and of their anti‑oncogenic effects. The findings presented herein lend further support to the antitumour effects of MKK6; MKK6 may thus represent a novel target for advanced personalised treatments against ERMS.
Collapse
Affiliation(s)
- Agnese Di Rocco
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Simona Camero
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, I‑00161 Rome, Italy
| | - Anna Benedetti
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics (AHFMO), Unit of Histology, Sapienza University of Rome, I‑00161 Rome, Italy
| | - Biliana Lozanoska-Ochser
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics (AHFMO), Unit of Histology, Sapienza University of Rome, I‑00161 Rome, Italy
| | - Francesca Megiorni
- Department of Experimental Medicine, Sapienza University of Rome, I‑00161 Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, I‑00161 Rome, Italy
| | - Lorenzo Stramucci
- Department of Diagnostic Research and Technological Innovation, IRCSS‑Regina Elena National Cancer Institute, I‑00144 Rome, Italy
| | - Carmela Ciccarelli
- Department of Life, Health and Environmental Sciences (MESVA), University of L'Aquila, I‑67100 L'Aquila, Italy
| | - Marina Bouché
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics (AHFMO), Unit of Histology, Sapienza University of Rome, I‑00161 Rome, Italy
| | - Gianluca Bossi
- Department of Diagnostic Research and Technological Innovation, IRCSS‑Regina Elena National Cancer Institute, I‑00144 Rome, Italy
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, I‑00161 Rome, Italy
| | - Bianca Maria Zani
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics (AHFMO), Unit of Histology, Sapienza University of Rome, I‑00161 Rome, Italy
| |
Collapse
|
4
|
Nicolas HA, Hua K, Quigley H, Ivare J, Tesson F, Akimenko MA. A CRISPR/Cas9 zebrafish lamin A/C mutant model of muscular laminopathy. Dev Dyn 2021; 251:645-661. [PMID: 34599606 DOI: 10.1002/dvdy.427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 08/13/2021] [Accepted: 09/16/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Lamin A/C gene (LMNA) mutations frequently cause cardiac and/or skeletal muscle diseases called striated muscle laminopathies. We created a zebrafish muscular laminopathy model using CRISPR/Cas9 technology to target the zebrafish lmna gene. RESULTS Heterozygous and homozygous lmna mutants present skeletal muscle damage at 1 day post-fertilization (dpf), and mobility impairment at 4 to 7 dpf. Cardiac structure and function analyses between 1 and 7 dpf show mild and transient defects in the lmna mutants compared to wild type (WT). Quantitative RT-PCR analysis of genes implicated in striated muscle laminopathies show a decrease in jun and nfκb2 expression in 7 dpf homozygous lmna mutants compared to WT. Homozygous lmna mutants have a 1.26-fold protein increase in activated Erk 1/2, kinases associated with striated muscle laminopathies, compared to WT at 7 dpf. Activated Protein Kinase C alpha (Pkc α), a kinase that interacts with lamin A/C and Erk 1/2, is also upregulated in 7 dpf homozygous lmna mutants compared to WT. CONCLUSIONS This study presents an animal model of skeletal muscle laminopathy where heterozygous and homozygous lmna mutants exhibit prominent skeletal muscle abnormalities during the first week of development. Furthermore, this is the first animal model that potentially implicates Pkc α in muscular laminopathies.
Collapse
Affiliation(s)
- Hannah A Nicolas
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, Ontario, Canada
| | - Khang Hua
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, Ontario, Canada
| | - Hailey Quigley
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, Ontario, Canada
| | - Joshua Ivare
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, Ontario, Canada
| | - Frédérique Tesson
- Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Marie-Andrée Akimenko
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
5
|
Ghosh S, Juin SK, Bhattacharyya Majumdar S, Majumdar S. Crucial role of glucosylceramide synthase in the regulation of stem cell-like cancer cells in B16F10 murine melanoma. Mol Carcinog 2021; 60:840-858. [PMID: 34516706 DOI: 10.1002/mc.23347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/15/2021] [Accepted: 08/25/2021] [Indexed: 11/06/2022]
Abstract
Cancer stem cells render a complex cascade of events that facilitates highly invasive melanoma malignancy. Interplay between immunocytes and cancer stem cells within tumor microenvironment with the participation of sphingolipid signaling mediators skews the immune evasion strategies toward metastatic neoplasm. In this context, we aimed to explore the functional aspect of glucosylceramide synthase (GCS), a key enzyme of sphingolipid biosynthesis in the maintenance of melanoma stem cell-like cancer cells (CSCs). Our findings demonstrated that tumor hypoxia was responsible for elevated GCS expression in melanoma, which was correlated with substantially increased melanoma CSCs. Moreover, hypoxia-induced TGF-β from TAMs and Tregs promoted GCS induction in B16F10 murine melanoma CSCs via PKCα signaling and facilitated the expansion of melanoma CSCs. Interestingly, GCS ablation hindered the immunosuppressiveness of TAMs and Tregs. Therefore, our study for the first time demonstrated a novel paracrine pathway of melanoma CSC maintenance and tumorigenicity, exploiting the bidirectional signaling with immunocytes. Furthermore, our study showed that the combinatorial immunotherapy involving immunomodulators like Mw and DTA-1 repressed CSC pool affecting GCS functions in advanced-stage B16F10 murine melanoma tumor. Moreover, GCS inhibition sensitized conventional chemotherapeutic drug-resistant melanoma CSCs to the genotoxic drugs paving the way toward selective melanoma treatment. Better therapeutic efficacy with inhibition of GCS and CSC depletion suggests a crucial role of GCS in melanoma treatment, therefore, implying its application concerning clinical challenges of chemotherapy resistance leading to prolonged survival.
Collapse
Affiliation(s)
- Sweta Ghosh
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | | | | |
Collapse
|
6
|
Zoroddu S, Marchesi I, Bagella L. PRC2: an epigenetic multiprotein complex with a key role in the development of rhabdomyosarcoma carcinogenesis. Clin Epigenetics 2021; 13:156. [PMID: 34372908 PMCID: PMC8351429 DOI: 10.1186/s13148-021-01147-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/02/2021] [Indexed: 02/02/2023] Open
Abstract
Skeletal muscle formation represents a complex of highly organized and specialized systems that are still not fully understood. Epigenetic systems underline embryonic development, maintenance of stemness, and progression of differentiation. Polycomb group proteins play the role of gene silencing of stemness markers that regulate muscle differentiation. Enhancer of Zeste EZH2 is the catalytic subunit of the complex that is able to trimethylate lysine 27 of histone H3 and induce silencing of the involved genes. In embryonal Rhabdomyosarcoma and several other tumors, EZH2 is often deregulated and, in some cases, is associated with tumor malignancy. This review explores the molecular processes underlying the failure of muscle differentiation with a focus on the PRC2 complex. These considerations could open new studies aimed at the development of new cutting-edge therapeutic strategies in the onset of Rhabdomyosarcoma.
Collapse
Affiliation(s)
- Stefano Zoroddu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100, Sassari, Italy
| | - Irene Marchesi
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100, Sassari, Italy
- Kitos Biotech Srls, Tramariglio, Alghero, SS, Italy
| | - Luigi Bagella
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100, Sassari, Italy.
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Yin YH, Zhang XH, Wang XA, Li RH, Zhang YW, Shan XX, You XX, Huang XD, Wu AL, Wang M, Pan XF, Bian C, Jiang WS, Shi Q, Yang JX. Construction of a chromosome-level genome assembly for genome-wide identification of growth-related quantitative trait loci in Sinocyclocheilus grahami (Cypriniformes, Cyprinidae). Zool Res 2021; 42:262-266. [PMID: 33764016 PMCID: PMC8175956 DOI: 10.24272/j.issn.2095-8137.2020.321] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The Dianchi golden-line barbel, Sinocyclocheilus grahami (Regan, 1904), is one of the “Four Famous Fishes” of Yunnan Province, China. Given its economic value, this species has been artificially bred successfully since 2007, with a nationally selected breed (“S. grahami, Bayou No. 1”) certified in 2018. For the future utilization of this species, its growth rate, disease resistance, and wild adaptability need to be improved, which could be achieved with the help of molecular marker-assisted selection (MAS). In the current study, we constructed the first chromosome-level genome of S. grahami, assembled 48 pseudo-chromosomes, and obtained a genome assembly of 1.49 Gb. We also performed QTL-seq analysis of S. grahami using the highest and lowest bulks (i.e., largest and smallest size) in both a sibling and random population. We screened two quantitative trait loci (QTLs) (Chr3, 14.9–39.1 Mb and Chr17, 4.1–27.4 Mb) as the major growth-related locations. Several candidate genes (e.g., map2k5, stat1, phf21a, sox6, and smad6) were also identified, with functions related to growth, such as cell differentiation, neuronal development, skeletal muscle development, chondrogenesis, and immunity. These results built a solid foundation for in-depth MAS studies on the growth traits of S. grahami.
Collapse
Affiliation(s)
- Yan-Hui Yin
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin-Hui Zhang
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, Guangdong 518083, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518083, China
| | - Xiao-Ai Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China
| | - Rui-Han Li
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, Guangdong 518083, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518083, China
| | - Yuan-Wei Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China
| | - Xin-Xin Shan
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, Guangdong 518083, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518083, China
| | - Xin-Xin You
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, Guangdong 518083, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518083, China
| | - Xin-Di Huang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - An-Li Wu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China
| | - Mo Wang
- Key Laboratory for Conserving Wildlife with Small Populations in Yunnan, Faculty of Biodiversity Conservation, Southwest Forestry University, Kunming, Yunnan 650224, China
| | - Xiao-Fu Pan
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China
| | - Chao Bian
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, Guangdong 518083, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518083, China
| | - Wan-Sheng Jiang
- Hunan Engineering Laboratory for Chinese Giant Salamander's Resource Protection and Comprehensive Utilization, and Key Laboratory of Hunan Forest and Chemical Industry Engineering, Jishou University, Zhangjiajie, Hunan 427000, China. E-mail:
| | - Qiong Shi
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, Guangdong 518083, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518083, China
| | - Jun-Xing Yang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China.,Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650224, China. E-mail:
| |
Collapse
|
8
|
Wojciechowicz T, Billert M, Dhandapani P, Szczepankiewicz D, Wasielewski O, Strowski MZ, Nowak KW, Skrzypski M. Neuropeptide B promotes proliferation and differentiation of rat brown primary preadipocytes. FEBS Open Bio 2021; 11:1153-1164. [PMID: 33629519 PMCID: PMC8016125 DOI: 10.1002/2211-5463.13128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/27/2021] [Accepted: 02/23/2021] [Indexed: 12/28/2022] Open
Abstract
Neuropeptide B (NPB) is reported to regulate energy homeostasis and metabolism via the NPBWR1 and NPBWR2 receptors in various tissues. However, the molecular mechanisms triggered from their interaction are not well investigated in brown adipose tissue. In this study, we specifically analyzed the role of NPB in controlling brown adipogenesis in rat brown preadipocytes. We first detected the expression of NPBWR1 and NPB on mRNA and protein level in brown preadipocytes and observed that NPB increased viability and proliferation of preadipocytes. Moreover, NPB stimulated expression of adipogenic genes (Prdm16, Ucp1) and suppressed the expression of antiadipogenic preadipocyte factor 1 (Pref1) during the differentiation process. Altogether, this led to an increase in intracellular lipid accumulation during preadipocyte differentiation, coupled with an increase in adrenaline‐induced oxygen consumption mediated by NPB. Furthermore, Ucp1 expression stimulated by NPB was attenuated by blockade of p38 kinase. In summary, we conclude that NPB promotes proliferation and differentiation of rat brown preadipocytes via p38‐dependent mechanism and plays an important role in controlling brown adipose tissue formation.
Collapse
Affiliation(s)
- Tatiana Wojciechowicz
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Poland.,Department of Hepatology and Gastroenterology, Charité-University Medicine Berlin, Germany
| | - Maria Billert
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Poland
| | - Priyavathi Dhandapani
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Poland
| | - Dawid Szczepankiewicz
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Poland
| | | | - Mathias Z Strowski
- Department of Hepatology and Gastroenterology, Charité-University Medicine Berlin, Germany.,Department of Internal Medicine-Gastroenterology & Oncology, Park-Klinik Weissensee, Berlin, Germany
| | - Krzysztof W Nowak
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Poland
| | - Marek Skrzypski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Poland
| |
Collapse
|
9
|
Xiang X, Langlois S, St-Pierre ME, Blinder A, Charron P, Graber TE, Fowler SL, Baird SD, Bennett SAL, Alain T, Cowan KN. Identification of pannexin 1-regulated genes, interactome, and pathways in rhabdomyosarcoma and its tumor inhibitory interaction with AHNAK. Oncogene 2021; 40:1868-1883. [PMID: 33564071 PMCID: PMC7946643 DOI: 10.1038/s41388-020-01623-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 01/31/2023]
Abstract
Rhabdomyosarcoma (RMS), the most common soft tissue sarcoma in children, is an aggressive cancer with a poor prognosis. Despite current management, the 5-year survival rate for patients with metastatic RMS is ∼30%; underscoring the need to develop better treatment strategies. We have recently reported that pannexin 1 (PANX1) levels are downregulated in RMS and that restoring its expression inhibits RMS progression. Here, we have surveyed and characterized the molecular changes induced by PANX1 re-expression in RMS. We cataloged transcriptomic changes in this context by RNA sequencing. At the protein level, we unveiled PANX1 interactors using BioID, complemented by co-immunoprecipitation coupled to high-performance liquid chromatography/electrospray ionization tandem mass spectrometry performed in PANX1-enriched fractions. Using these data, we generated searchable public databases for the PANX1 interactome and changes to the RMS transcriptome occurring when PANX1 expression is restored. STRING network analyses revealed a PANX1 interactome involving plasma membrane and cytoskeleton-associated proteins including the previously undescribed interactor AHNAK. Indeed, AHNAK knockdown abrogated the PANX1-mediated reduction in RMS cell viability and migration. Using these unbiased approaches, we bring insight to the mechanisms by which PANX1 inhibits RMS progression, identifying the cell migration protein AHNAK as a key modifier of PANX1-mediated changes in RMS malignant properties.
Collapse
Affiliation(s)
- Xiao Xiang
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Stéphanie Langlois
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Surgery, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada
| | - Marie-Eve St-Pierre
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Anna Blinder
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Philippe Charron
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Tyson E Graber
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Stephanie L Fowler
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Neural Regeneration Laboratory and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
- UK Dementia Research Institute, University College London, London, UK
| | - Stephen D Baird
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Steffany A L Bennett
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Neural Regeneration Laboratory and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Tommy Alain
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Kyle N Cowan
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
- Department of Surgery, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
10
|
Radzikowska J, Krzeski A, Czarnecka AM, Klepacka T, Rychlowska-Pruszynska M, Raciborska A, Dembowska-Baginska B, Pronicki M, Kukwa A, Sierdzinski J, Kukwa W. Endoglin Expression and Microvessel Density as Prognostic Factors in Pediatric Rhabdomyosarcoma. J Clin Med 2021; 10:jcm10030512. [PMID: 33535525 PMCID: PMC7867094 DOI: 10.3390/jcm10030512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/17/2021] [Accepted: 01/23/2021] [Indexed: 12/23/2022] Open
Abstract
(1) Background: The study proposed to analyze microvessel density (MVD) in rhabdomyosarcoma (RMS) based on the expression of angiogenesis markers and define its prognostic role in this group of patients. (2) Methods: The study included forty-nine pediatric patients diagnosed with RMS. Tumor tissue expression of CD31, CD34, and CD105 was analyzed. MVD was calculated and correlated with clinical RMS prognostic parameters. (3) Results: CD31, CD34, and CD105 are expressed in all RMS cases. MVD/CD105 was significantly higher in the RMS group than in the control group. The mean and median values of MVD/CD105 in RMS were lower than MVD/CD31 and MVD/CD34. MVD/CD105 was significantly higher in patients with alveolar RMS and those with metastatic disease. Patients with higher levels of MVD/CD105 had a higher risk of death (HR = 1.009). (4) Conclusion: CD105 is a relevant angiogenesis marker in pediatric RMS, and MVD/CD105 is an independent risk factor of short overall survival in children with RMS.
Collapse
Affiliation(s)
- Joanna Radzikowska
- Department of Otorhinolaryngology, Faculty of Dental Medicine, Medical University of Warsaw, 19/25 Stepinska St., 00-739 Warsaw, Poland; (J.R.); (A.K.)
| | - Antoni Krzeski
- Department of Otorhinolaryngology, Faculty of Dental Medicine, Medical University of Warsaw, 19/25 Stepinska St., 00-739 Warsaw, Poland; (J.R.); (A.K.)
| | - Anna M. Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute—Oncology Center, 5 Roentgena St., 02-781 Warsaw, Poland;
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego St., 02-106 Warsaw, Poland
| | - Teresa Klepacka
- Department of Pathology, Institute of Mother and Child, 17a Kasprzaka St., 01-211 Warsaw, Poland;
| | - Magdalena Rychlowska-Pruszynska
- Department of Oncology and Surgical Oncology for Children and Youth, Institute of Mother and Child, 17a Kasprzaka St., 01-211 Warsaw, Poland; (M.R.-P.); (A.R.)
| | - Anna Raciborska
- Department of Oncology and Surgical Oncology for Children and Youth, Institute of Mother and Child, 17a Kasprzaka St., 01-211 Warsaw, Poland; (M.R.-P.); (A.R.)
| | - Bozenna Dembowska-Baginska
- Department of Pediatric Oncology, The Children’s Memorial Health Institute, 20 Dzieci Polskich St., 04-730 Warsaw, Poland;
| | - Maciej Pronicki
- Department of Pathology, The Children’s Memorial Health Institute, 20 Dzieci Polskich St., 04-730 Warsaw, Poland;
| | - Andrzej Kukwa
- Department of Otolaryngology and Head and Neck Diseases, School of Medicine, University of Warmia and Mazury, 30 Warszawska St., 10-082 Olsztyn, Poland;
| | - Janusz Sierdzinski
- Department of Medical Informatics and Telemedicine, Medical University of Warsaw, 14/16 Litewska St., 00-581 Warsaw, Poland;
| | - Wojciech Kukwa
- Department of Otorhinolaryngology, Faculty of Dental Medicine, Medical University of Warsaw, 19/25 Stepinska St., 00-739 Warsaw, Poland; (J.R.); (A.K.)
- Correspondence: ; Tel.: +48-223186270
| |
Collapse
|
11
|
Nicolas HA, Bertrand AT, Labib S, Mohamed-Uvaize M, Bolongo PM, Wu WY, Bilińska ZT, Bonne G, Akimenko MA, Tesson F. Protein Kinase C Alpha Cellular Distribution, Activity, and Proximity with Lamin A/C in Striated Muscle Laminopathies. Cells 2020; 9:cells9112388. [PMID: 33142761 PMCID: PMC7693451 DOI: 10.3390/cells9112388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/19/2020] [Accepted: 10/28/2020] [Indexed: 11/24/2022] Open
Abstract
Striated muscle laminopathies are cardiac and skeletal muscle conditions caused by mutations in the lamin A/C gene (LMNA). LMNA codes for the A-type lamins, which are nuclear intermediate filaments that maintain the nuclear structure and nuclear processes such as gene expression. Protein kinase C alpha (PKC-α) interacts with lamin A/C and with several lamin A/C partners involved in striated muscle laminopathies. To determine PKC-α’s involvement in muscular laminopathies, PKC-α’s localization, activation, and interactions with the A-type lamins were examined in various cell types expressing pathogenic lamin A/C mutations. The results showed aberrant nuclear PKC-α cellular distribution in mutant cells compared to WT. PKC-α activation (phos-PKC-α) was decreased or unchanged in the studied cells expressing LMNA mutations, and the activation of its downstream targets, ERK 1/2, paralleled PKC-α activation alteration. Furthermore, the phos-PKC-α-lamin A/C proximity was altered. Overall, the data showed that PKC-α localization, activation, and proximity with lamin A/C were affected by certain pathogenic LMNA mutations, suggesting PKC-α involvement in striated muscle laminopathies.
Collapse
Affiliation(s)
- Hannah A. Nicolas
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (H.A.N.); (W.Y.W.); (M.-A.A.)
| | - Anne T. Bertrand
- Sorbonne Université, Inserm, Centre de Recherche en Myologie, UMRS 974, G.H. Pitié-Salpêtrière, 75013 Paris, France; (A.T.B.); (G.B.)
| | - Sarah Labib
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (S.L.); (M.M.-U.); (P.M.B.)
| | - Musfira Mohamed-Uvaize
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (S.L.); (M.M.-U.); (P.M.B.)
| | - Pierrette M. Bolongo
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (S.L.); (M.M.-U.); (P.M.B.)
| | - Wen Yu Wu
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (H.A.N.); (W.Y.W.); (M.-A.A.)
| | - Zofia T. Bilińska
- Unit for Screening Studies in Inherited Cardiovascular Diseases, National Institute of Cardiology, 04-628 Warsaw, Poland;
| | - Gisèle Bonne
- Sorbonne Université, Inserm, Centre de Recherche en Myologie, UMRS 974, G.H. Pitié-Salpêtrière, 75013 Paris, France; (A.T.B.); (G.B.)
| | - Marie-Andrée Akimenko
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (H.A.N.); (W.Y.W.); (M.-A.A.)
| | - Frédérique Tesson
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (S.L.); (M.M.-U.); (P.M.B.)
- Correspondence: ; Tel.: +1-613-562-5800 (ext. 7370)
| |
Collapse
|
12
|
Li JY, Chen RJ, Huang LT, Lee TY, Lu WJ, Lin KH. Embelin as a Novel Inhibitor of PKC in the Prevention of Platelet Activation and Thrombus Formation. J Clin Med 2019; 8:jcm8101724. [PMID: 31635287 PMCID: PMC6832570 DOI: 10.3390/jcm8101724] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/09/2019] [Accepted: 10/16/2019] [Indexed: 12/25/2022] Open
Abstract
Embelin is a quinone derivative and found in the fruits of Embelia ribes Burm.f. Embelin has been identified as a small molecular inhibitor of X-chromosome-linked inhibitor of apoptosis proteins, and has multiple biological activities, including antioxidation, anti-inflammation, and antitumor effects. However, the effect of embelin in platelets remains unclear. Thus, this study investigated the antiplatelet mechanism of embelin. Our data revealed that embelin could inhibit platelet aggregation induced by various agonists, including the protein kinase C (PKC) activator phorbol 12,13-dibutyrate (PDBu). Embelin, as well as the PKC inhibitor Ro 31-8220, markedly reduced PDBu-mediated phosphorylation of the PKC substrate, suggesting that embelin may be a PKC inhibitor for platelets. Embelin could block PKC downstream signaling and events, including the inhibition of protein kinase B and mitogen-activated protein kinase activation, granule release, and glycoprotein IIbIIIa activation. Moreover, embelin could delay thrombus formation in the mesenteric microvessels of mice, but did not significantly affect the tail bleeding time. In conclusion, we demonstrated that embelin is a PKC inhibitor and possesses antiplatelet and antithrombotic effects. The further analysis is necessary to more accurately determine clinical therapeutic potential of embelin in all clinical thromboembolic events with disturbance of thrombocyte function.
Collapse
Affiliation(s)
- Jiun Yi Li
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan.
- Department of Surgery, MacKay Memorial Hospital, Taipei 104, Taiwan.
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Ray Jade Chen
- Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Li Ting Huang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Tzu Yin Lee
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Wan Jung Lu
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan.
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan.
| | - Kuan Hung Lin
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 252, Taiwan.
| |
Collapse
|
13
|
Takashima I, Kusamori K, Hakariya H, Takashima M, Vu TH, Mizukami Y, Noda N, Takayama Y, Katsuda Y, Sato SI, Takakura Y, Nishikawa M, Uesugi M. Multifunctionalization of Cells with a Self-Assembling Molecule to Enhance Cell Engraftment. ACS Chem Biol 2019; 14:775-783. [PMID: 30807095 DOI: 10.1021/acschembio.9b00109] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cell-based therapy is a promising approach to restoring lost functions to compromised organs. However, the issue of inefficient cell engraftment remains to be resolved. Herein, we take a chemical approach to facilitate cell engraftment by using self-assembling molecules which modify two cellular traits: cell survival and invasiveness. In this system, the self-assembling molecule induces syndecan-4 clusters on the cellular surface, leading to enhanced cell viability. Further integration with Halo-tag technology provided this self-assembly structure with matrix metalloproteinase-2 to functionalize cells with cell-invasion activity. In vivo experiments showed that the pretreated cells were able to survive injection and then penetrate and engraft into the host tissue, demonstrating that the system enhances cell engraftment. Therefore, cell-surface modification via an alliance between self-assembling molecules and ligation technologies may prove to be a promising method for cell engraftment.
Collapse
Affiliation(s)
- Ippei Takashima
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Kosuke Kusamori
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Hayase Hakariya
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Megumi Takashima
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Thi Hue Vu
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yuya Mizukami
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Naotaka Noda
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yukiya Takayama
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Yousuke Katsuda
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Shin-ichi Sato
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yoshinobu Takakura
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Makiya Nishikawa
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba 278-8510, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Motonari Uesugi
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Uji, Kyoto 611-0011, Japan
- School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
14
|
Khamo JS, Krishnamurthy VV, Chen Q, Diao J, Zhang K. Optogenetic Delineation of Receptor Tyrosine Kinase Subcircuits in PC12 Cell Differentiation. Cell Chem Biol 2018; 26:400-410.e3. [PMID: 30595532 DOI: 10.1016/j.chembiol.2018.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 08/13/2018] [Accepted: 11/02/2018] [Indexed: 01/19/2023]
Abstract
Nerve growth factor elicits signaling outcomes by interacting with both its high-affinity receptor, TrkA, and its low-affinity receptor, p75NTR. Although these two receptors can regulate distinct cellular outcomes, they both activate the extracellular-signal-regulated kinase pathway upon nerve growth factor stimulation. To delineate TrkA subcircuits in PC12 cell differentiation, we developed an optogenetic system whereby light was used to specifically activate TrkA signaling in the absence of nerve growth factor. By using tyrosine mutants of the optogenetic TrkA in combination with pathway-specific pharmacological inhibition, we find that Y490 and Y785 each contributes to PC12 cell differentiation through the extracellular-signal-regulated kinase pathway in an additive manner. Optogenetic activation of TrkA eliminates the confounding effect of p75NTR and other potential off-target effects of the ligand. This approach can be generalized for the mechanistic study of other receptor-mediated signaling pathways.
Collapse
Affiliation(s)
- John S Khamo
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Vishnu V Krishnamurthy
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Qixin Chen
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| | - Kai Zhang
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
15
|
Raja R, Lata S, Trivedi S, Banerjea AC. Serum deprivation/starvation leads to reactivation of HIV-1 in latently infected monocytes via activating ERK/JNK pathway. Sci Rep 2018; 8:14496. [PMID: 30262819 PMCID: PMC6160481 DOI: 10.1038/s41598-018-32316-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/03/2018] [Indexed: 01/04/2023] Open
Abstract
Despite the high success rate, antiretroviral therapy does not cure the disease completely due to presence of latent viral reservoirs. Although several studies have addressed this issue earlier, the role of serum starvation/deprivation in HIV-1 latency has not been studied. So, we investigated the role of serum starvation in regulating HIV-1 latency. The impact of serum starvation on HIV-1 latency was assessed in latently infected monocytes U1 and T-cells J1.1. Serum starvation breaks HIV-1 latency in U1 cells. Under similar conditions, J1.1 cells failed to show reactivation of virus. We investigated the involvement of cell death pathway and autophagy during the serum starvation in viral reactivation. Inhibition of these pathways did not affect viral reactivation. Furthermore, other crucial factors like NF-κB, SP1 and AKT did not play any role in regulating viral latency. Here, we report that serum deprivation up-regulates ERK/JNK pathway. This leads to phosphorylation of c-Jun which plays an important role in viral reactivation. Treatment of cells with U0126, an ERK kinase inhibitor, potently inhibited viral replication. In summary, we show that serum starvation leads to reactivation of HIV-1 in latently infected monocytes through the ERK/JNK pathway.
Collapse
Affiliation(s)
- Rameez Raja
- Laboratory of Virology, National Institute of Immunology, New Delhi, India.,Lerner Research Institute, Cleveland Clinic, Ohio, USA
| | - Sneh Lata
- Laboratory of Virology, National Institute of Immunology, New Delhi, India
| | - Shubhendu Trivedi
- Laboratory of Virology, National Institute of Immunology, New Delhi, India
| | - Akhil C Banerjea
- Laboratory of Virology, National Institute of Immunology, New Delhi, India.
| |
Collapse
|
16
|
Codenotti S, Poli M, Asperti M, Zizioli D, Marampon F, Fanzani A. Cell growth potential drives ferroptosis susceptibility in rhabdomyosarcoma and myoblast cell lines. J Cancer Res Clin Oncol 2018; 144:1717-1730. [PMID: 29971532 DOI: 10.1007/s00432-018-2699-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 06/29/2018] [Indexed: 12/16/2022]
Abstract
PURPOSE Ferroptosis is a programmed form of iron-dependent cell death caused by lipid hydroperoxide accumulation, which can be prevented by glutathione peroxidase 4 (GPx4) activity. Here we investigated the effects of ferroptosis inducers called erastin and RSL3, which act by glutathione depletion and GPx4 inactivation, respectively, on muscle-derived cell lines of embryonal and alveolar rhabdomyosarcoma (RMS), and mouse normal skeletal C2C12 myoblasts. METHODS Myogenic lines were exposed to stepwise increasing concentrations of ferroptosis inducers either alone or in combination with iron supplementation, iron chelating agents (bathophenanthrolinedisulfonic acid, BPS), antioxidant molecules (glutathione, N-acetylcysteine), lipid peroxidation inhibitors (ferrostatin-1), and chemotherapeutic agents (doxorubicin and actinomycin D). Drug susceptibility was quantified by measuring cell viability, proliferation and differentiation via neutral red assay, crystal violet assay and Giemsa staining, respectively. The detection of lipid hydroperoxide and protein levels was performed by immunofluorescence and Western blot analysis, respectively. RESULTS Erastin and RSL3 increased lipid hydroperoxide levels preferentially in the embryonal U57810 and myoblast C2C12 lines, leading to ferroptosis that was accentuated by iron supplementation or prevented by co-treatment with BPS, glutathione, N-acetylcysteine and ferrostatin-1. The inhibition of extracellular regulated kinases (ERK) pathway prevented ferroptosis in U57810 and C2C12 cells, whereas its increased activation in the embryonal RD cells mediated by caveolin-1 (Cav-1) overexpression led to augmented ferroptosis susceptibility. Finally, we observed the combination of erastin or RSL3 with chemotherapeutic doxorubicin and actinomycin D agents to be effective in increasing cell death in all RMS lines. CONCLUSIONS Erastin and RSL3 trigger ferroptosis in highly proliferating myogenic lines through a ERK pathway-dependent fashion.
Collapse
Affiliation(s)
- Silvia Codenotti
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Maura Poli
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Michela Asperti
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Daniela Zizioli
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesco Marampon
- Division of Radiation Oncology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alessandro Fanzani
- Division of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
17
|
Amiri M, Yousefnia S, Seyed Forootan F, Peymani M, Ghaedi K, Nasr Esfahani MH. Diverse roles of fatty acid binding proteins (FABPs) in development and pathogenesis of cancers. Gene 2018; 676:171-183. [PMID: 30021130 DOI: 10.1016/j.gene.2018.07.035] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 07/04/2018] [Accepted: 07/12/2018] [Indexed: 12/27/2022]
Abstract
One of the most importantly involved pathways in cancer development is fatty-acid signaling pathway. Synthesized lipids as energetic sources are consumed by cancer cells for proliferation, growth, survival, invasion and angiogenesis. Fatty acids as signaling compounds regulate metabolic and transcriptional networks, survival pathways and inflammatory responses. Aggregation of fatty acids with fatty acid binding proteins (FABPs) facilitates their transportation to different cell organelles. FABPs, a group of lipid binding proteins modulate fatty acid metabolism, cell growth and proliferation and cancer development. They may be used as tumor marker in some cancers. FABPs are expressed in most malignancies such as prostate, breast, liver, bladder and lung cancer which are associated with the incidence, proliferation, metastasis, invasion of tumors. This review introduces several isoforms of FABPs (FABP1-12) and summarizes their function and their possible roles in cancer development through some proposed mechanisms.
Collapse
Affiliation(s)
- Mina Amiri
- Department of Modern Biology, ACECR Institute of Higher Education (Isfahan Branch), Isfahan, Iran
| | - Saghar Yousefnia
- Division of Cellular and Molecular Biology, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Farzad Seyed Forootan
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran; Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran.
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran; Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Kamran Ghaedi
- Division of Cellular and Molecular Biology, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran; Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| | - Mohammad Hossein Nasr Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
18
|
Takami M, Katayama K, Noguchi K, Sugimoto Y. Protein kinase C alpha-mediated phosphorylation of PIM-1L promotes the survival and proliferation of acute myeloid leukemia cells. Biochem Biophys Res Commun 2018; 503:1364-1371. [PMID: 30017192 DOI: 10.1016/j.bbrc.2018.07.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 11/27/2022]
Abstract
FMS-like tyrosine kinase 3 (FLT3)-internal tandem duplication (ITD) is a constitutively active mutant of FLT3 and causes 20%-30% of acute myeloid leukemia (AML) cases. FLT3-ITD upregulates the proviral integration site for Moloney murine leukemia virus 1 (PIM-1) expression and promotes the proliferation of AML cells. In this study, we investigated the role of protein kinase C (PKC)-mediated phosphorylation on the expression and function of PIM-1L. Drug screening in leukemia cell lines revealed that sotrastaurin (a PKC inhibitor) suppressed the proliferation of the FLT3-ITD-positive AML cell line MV4-11 but not of K562, HL60, or KG-1a cells, similar to SGI-1776 (a PIM-1/FLT3 inhibitor) and quizartinib (an FLT3 inhibitor). Sotrastaurin decreased the expression of pro-survival protein myeloid cell leukemia (MCL-1) and the phosphorylation of eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), both of which are downstream effectors of PIM-1. PKCα directly phosphorylated Ser65 of PIM-1L, which is a long isoform of PIM-1. The PKCα-mediated phosphorylation stabilized PIM-1L. The phosphorylation-mimicked mutant, PIM-1L-S65D, was more stable and showed higher kinase activity than PIM-1L-S65A. Expression of PIM-1L-wildtype or -S65D reduced sotrastaurin-mediated apoptosis and growth inhibition in MV4-11 transfectants. These results suggest that PKCα directly upregulates PIM-1L, resulting in promotion of the survival and proliferation of AML cells.
Collapse
Affiliation(s)
- Mayu Takami
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Kazuhiro Katayama
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, Tokyo, Japan.
| | - Kohji Noguchi
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Yoshikazu Sugimoto
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, Tokyo, Japan
| |
Collapse
|
19
|
Biomaterials in Tendon and Skeletal Muscle Tissue Engineering: Current Trends and Challenges. MATERIALS 2018; 11:ma11071116. [PMID: 29966303 PMCID: PMC6073924 DOI: 10.3390/ma11071116] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/20/2018] [Accepted: 06/25/2018] [Indexed: 12/17/2022]
Abstract
Tissue engineering is a promising approach to repair tendon and muscle when natural healing fails. Biohybrid constructs obtained after cells’ seeding and culture in dedicated scaffolds have indeed been considered as relevant tools for mimicking native tissue, leading to a better integration in vivo. They can also be employed to perform advanced in vitro studies to model the cell differentiation or regeneration processes. In this review, we report and analyze the different solutions proposed in literature, for the reconstruction of tendon, muscle, and the myotendinous junction. They classically rely on the three pillars of tissue engineering, i.e., cells, biomaterials and environment (both chemical and physical stimuli). We have chosen to present biomimetic or bioinspired strategies based on understanding of the native tissue structure/functions/properties of the tissue of interest. For each tissue, we sorted the relevant publications according to an increasing degree of complexity in the materials’ shape or manufacture. We present their biological and mechanical performances, observed in vitro and in vivo when available. Although there is no consensus for a gold standard technique to reconstruct these musculo-skeletal tissues, the reader can find different ways to progress in the field and to understand the recent history in the choice of materials, from collagen to polymer-based matrices.
Collapse
|
20
|
Cheng Y, Zhu Y, Xu W, Xu J, Yang M, Chen P, Zhao J, Geng L, Gong S. PKCα in colon cancer cells promotes M1 macrophage polarization via MKK3/6-P38 MAPK pathway. Mol Carcinog 2018; 57:1017-1029. [PMID: 29637628 DOI: 10.1002/mc.22822] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 03/17/2018] [Accepted: 04/04/2018] [Indexed: 01/19/2023]
Abstract
Tumor associated macrophages are potential targets of the immune therapy for patients with colon cancer. PKCα acts as a tumor suppressor in the intestine. However, the correlation between PKCα expressed in colon cancer cells and tumor associated macrophages polarization has never been detected. In the present study, the correlation between PKCα expression and level of M1 macrophages was evaluated in human colon cancer tissues. A xenograft mouse model of colon cancer cells with different PKCα expression level was constructed to evaluate the effect of PKCα on M1 macrophages polarization in vivo. Co-culture of colon cancer cells and differentiated macrophages was used to detect the potential interplay in vitro. PKCα regulated production of cytokines which correlated with macrophage polarization and the underlying mechanism was further explored. Our study showed that high PKCα expression in human colon cancer tissues correlated with better prognosis and high M1 macrophage content. PKCα expressed in colon cancer cells inhibited the growth of colon cancer in mice model. PKCα induced macrophages polarized to the M1-like phenotype both in vitro and in vivo. Mechanistically, PKCα targeted P38 via MKK3/6 to promote IL12 and GM-CSF expression which further enhanced M1-like macrophages polarization. In conclusion, this study provided evidence for the first time that PKCα in colon cancer cells play an anticancer action by inducing the polarization of tumor associated macrophages to M1-like phenotype in the tumor microenvironment. PKCα promoted IL12/GM-CSF-mediated M1 polarization through MKK3/6-P38 signaling pathway. Our investigation suggested that modulation of the PKCα signaling pathway might serve as a novel strategy for colon cancer therapy.
Collapse
Affiliation(s)
- Yang Cheng
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yun Zhu
- Liver Tumor Center, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wanfu Xu
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiajia Xu
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Min Yang
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Peiyu Chen
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Junhong Zhao
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lanlan Geng
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Sitang Gong
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
21
|
Ábrigo J, Campos F, Simon F, Riedel C, Cabrera D, Vilos C, Cabello-Verrugio C. TGF-β requires the activation of canonical and non-canonical signalling pathways to induce skeletal muscle atrophy. Biol Chem 2017; 399:253-264. [DOI: 10.1515/hsz-2017-0217] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/03/2017] [Indexed: 12/20/2022]
Abstract
Abstract
The transforming growth factor type-beta (TGF-β) induces skeletal muscle atrophy characterised by a decrease in the fibre’s diameter and levels of myosin heavy chain (MHC), also as an increase of MuRF-1 expression. In addition, TGF-β induces muscle atrophy by a mechanism dependent on reactive oxygen species (ROS). TGF-β signals by activating both canonical Smad-dependent, and non-canonical signalling pathways such as ERK1/2, JNK1/2, and p38 MAPKs. However, the participation of canonical and non-canonical signalling pathways in the TGF-β atrophic effect on skeletal muscle is unknown. We evaluate the impact of Smad and MAPK signalling pathways on the TGF-β-induced atrophic effect in C2C12 myotubes. The results indicate that TGF-β activates Smad2/3, ERK1/2 and JNK1/2, but not p38 in myotubes. The pharmacological inhibition of Smad3, ERK1/2 and JNK1/2 activation completely abolished the atrophic effect of TGF-β. Finally, the inhibition of these canonical and non-canonical pathways did not decrease the ROS increment, while the inhibition of ROS production entirely abolished the phosphorylation of Smad3, ERK1/2 and JNK1/2. These results suggest that TGF-β requires Smad3, ERK1/2 and JNK1/2 activation to produce skeletal muscle atrophy. Moreover, the induction of ROS by TGF-β is an upstream event to canonical and non-canonical pathways.
Collapse
Affiliation(s)
- Johanna Ábrigo
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Fabian Campos
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Felipe Simon
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Claudia Riedel
- Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas and Facultad de Medicina, Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
| | - Daniel Cabrera
- Universidad Bernardo O Higgins, Facultad de Salud , Departamento de Ciencias Químicas y Biológicas , 8370993 Santiago , Chile
- Departamento de Gastroenterología, Facultad de Medicina , Pontificia Universidad Católica de Chile , 8331150 Santiago , Chile
| | - Cristian Vilos
- Laboratory of Nanomedicine and Targeted Delivery, Center for Integrative Medicine and Innovative Science, Faculty of Medicine, and Center for Bioinformatics and Integrative Biology, Faculty of Biological Sciences , Universidad Andres Bello , 8370146 Santiago , Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA) , Universidad de Santiago de Chile , 9170022 Santiago , Chile
| | - Claudio Cabello-Verrugio
- Millennium Institute on Immunology and Immunotherapy , 8331150 Santiago , Chile
- Laboratory of Muscle Pathology, Fragility and Aging , Departmento de Ciencias Biológicas, Facultad de Ciencias Biológicas and Facultad de Medicina , Universidad Andres Bello , Avenida República 239 , Santiago 8370146 , Chile
| |
Collapse
|
22
|
Marchesi I, Sanna L, Fais M, Fiorentino FP, Giordano A, Bagella L. 12-O-tetradecanoylphorbol-13-acetate and EZH2 inhibition: A novel approach for promoting myogenic differentiation in embryonal rhabdomyosarcoma cells. J Cell Physiol 2017; 233:2360-2365. [DOI: 10.1002/jcp.26107] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 01/26/2023]
Affiliation(s)
- Irene Marchesi
- Department of Biomedical Sciences; University of Sassari; Sassari Italy
| | - Luca Sanna
- Department of Biomedical Sciences; University of Sassari; Sassari Italy
| | - Milena Fais
- Department of Biomedical Sciences; University of Sassari; Sassari Italy
| | | | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine; Center for Biotechnology; College of Science and Technology; Temple University; Philadelphia Pennsylvania
- Department of Medicine; Surgery and Neuroscience; University of Siena; Siena Italy
| | - Luigi Bagella
- Department of Biomedical Sciences; University of Sassari; Sassari Italy
- Sbarro Institute for Cancer Research and Molecular Medicine; Center for Biotechnology; College of Science and Technology; Temple University; Philadelphia Pennsylvania
| |
Collapse
|
23
|
Zhu Y, Tan YQ, Wang CC, Leung LK. The flame retardant 2,2',4,4'-Tetrabromodiphenyl ether enhances the expression of corticotropin-releasing hormone in the placental cell model JEG-3. CHEMOSPHERE 2017; 174:499-505. [PMID: 28189027 DOI: 10.1016/j.chemosphere.2017.01.144] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/23/2017] [Accepted: 01/30/2017] [Indexed: 06/06/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) are chemicals used as flame retardants in household products. After disposing of these items, PBDEs leach from the products by surface water. BDE-47 is a PBDE congener commonly isolated from contaminated food and is the most studied isomer. The placenta is the major source of hormones during pregnancy, and an elevated level of corticotrophin-releasing hormone (CRH) is associated with premature delivery. In the present study, we examined changes in the placental CRH expression under BDE-47 exposure in the JEG-3 cell model system. These placental cells are derived from human choriocarcinoma. Our result showed that this pollutant induced the CRH mRNA expression at 0.5 nM or above in the cells. A similar trend was observed when CRH peptide was determined by Western analysis in the cell lysates. As previous studies have shown the importance of signal transduction pathways in the gene regulation, the status of some protein kinases in the present study was investigated. The phosphorylated PKCα, JNK, and P38 were increased by the toxicant treatment, and administering the specific inhibitors could counteract the induced CRH expression. It appeared that the signaling transduction pathway of PKC was a significant contributor in the transcriptional regulation. Further study by using Electrophoretic Mobility Shift Assay suggested that AP-2 was the ultimate DNA-binding element for the initiation of gene transcription. Because an untimely increased CRH may compromise fetal development and induce preterm birth, the present study suggested that endocrine changes in pregnancy should be taken into consideration in the next assessment.
Collapse
Affiliation(s)
- Yun Zhu
- Food and Nutritional Sciences Programme, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| | - Yan Qin Tan
- Food and Nutritional Sciences Programme, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, Li Ka Shing Institute of Health Sciences and School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| | - Lai K Leung
- Food and Nutritional Sciences Programme, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong; Biochemistry Programme, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong.
| |
Collapse
|
24
|
Chen TH, Chen CY, Wen HC, Chang CC, Wang HD, Chuu CP, Chang CH. YAP promotes myogenic differentiation via the MEK5-ERK5 pathway. FASEB J 2017; 31:2963-2972. [PMID: 28356344 DOI: 10.1096/fj.201601090r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/13/2017] [Indexed: 01/26/2023]
Abstract
Yes-associated protein (YAP) is a transcriptional coactivator in the Hippo pathway that regulates cell proliferation, differentiation, and apoptosis. The MEK5/ERK5 MAPK cascade is essential for the early step of myogenesis. In this study, we generated C2C12 stable cell lines that expressed YAP (C2C12-YAP cells) and found that ERK5 and MEK5 were activated in C2C12-YAP cells compared with control C2C12 (C2C12-vector) cells. C2C12-YAP stable cells also differentiated into myotubes better than C2C12-vector cells, and expressed elevated levels of myogenin, a transcription factor that regulates myogenesis, as well as elevated levels of myosin heavy chain, a skeletal muscle marker. Western blot analysis revealed that Src and c-Abl (Abelson murine leukemia viral oncogene homolog 1) activation were enhanced in C2C12-YAP cells. Conversely, treatment of inhibitors of c-Abl, Src, or MEK5 inhibited activation of MEK5 and ERK5 and myogenesis of C2C12 myoblasts. Specific interactions between YAP and proteins in the ERK5 pathway, such as MEK kinase 3 (MEKK3) and ERK5, were illustrated by coimmunoprecipitation experiments. MEKK3 contains the PPGY motif (aa 178-181), which may interact with YAP. Site-directed mutagenesis experiments revealed that expression of MEKK3 Y181F mutant inhibited MEK5/ERK5 activation and myogenic differentiation. These results suggest that YAP promotes muscle differentiation by activating the Abl/Src/MEKK3/MEK5/ERK5 kinase cascade.-Chen, T.-H., Chen, C.-Y., Wen, H.-C., Chang, C.-C., Wang, H.-D., Chuu, C.-P., Chang, C.-H. YAP promotes myogenic differentiation via the MEK5-ERK5 pathway.
Collapse
Affiliation(s)
- Ting-Huan Chen
- Department of Life Science, National Tsing Hua University, Hsin-Chu, Taiwan, China.,Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, China
| | - Chen-Yu Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, China
| | - Hui-Chin Wen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, China
| | - Chia-Chu Chang
- Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan, China
| | - Horng-Dar Wang
- Department of Life Science, National Tsing Hua University, Hsin-Chu, Taiwan, China
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, China;
| | - Chung-Ho Chang
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, China; .,Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan, China
| |
Collapse
|
25
|
Megiorni F, Camero S, Ceccarelli S, McDowell HP, Mannarino O, Marampon F, Pizer B, Shukla R, Pizzuti A, Marchese C, Clerico A, Dominici C. DNMT3B in vitro knocking-down is able to reverse embryonal rhabdomyosarcoma cell phenotype through inhibition of proliferation and induction of myogenic differentiation. Oncotarget 2016; 7:79342-79356. [PMID: 27764816 PMCID: PMC5346718 DOI: 10.18632/oncotarget.12688] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/29/2016] [Indexed: 01/08/2023] Open
Abstract
Aberrant DNA methylation has been frequently observed in many human cancers, including rhabdomyosarcoma (RMS), the most common soft tissue sarcoma in children. To date, the expression and function of the de novo DNA methyltransferase (DNMT) 3B in RMS have not yet been investigated. Our study show for the first time a significant up-regulation of DNMT3B levels in 14 RMS tumour samples and 4 RMS cell lines in comparison to normal skeletal muscle. Transfection of RD and TE671 cells, two in vitro models of embryonal RMS (ERMS), with a synthetic DNMT3B siRNA decreased cell proliferation by arresting cell cycle at G1 phase, as demonstrated by the reduced expression of Cyclin B1, Cyclin D1 and Cyclin E2, and by the concomitant up-regulation of the checkpoint regulators p21 and p27. DNMT3B depletion also impaired RB phosphorylation status and decreased migratory capacity and clonogenic potential. Interestingly, DNMT3B knock-down was able to commit ERMS cells towards myogenic terminal differentiation, as confirmed by the acquisition of a myogenic-like phenotype and by the increased expression of the myogenic markers MYOD1, Myogenin and MyHC. Finally, inhibition of MEK/ERK signalling by U0126 resulted in a reduction of DNMT3B protein, giving evidence that DNMT3B is a down-stream molecule of this oncogenic pathway.Taken together, our data indicate that altered expression of DNMT3B plays a key role in ERMS development since its silencing is able to reverse cell cancer phenotype by rescuing myogenic program. Epigenetic therapy, by targeting the DNA methylation machinery, may represent a novel therapeutic strategy against RMS.
Collapse
Affiliation(s)
- Francesca Megiorni
- Department of Paediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Simona Camero
- Department of Paediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Simona Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Heather P. McDowell
- Department of Paediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
- Department of Oncology, Alder Hey Children's NHS Foundation Trust, Liverpool, United Kingdom
| | - Olga Mannarino
- Department of Paediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Francesco Marampon
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Barry Pizer
- Department of Oncology, Alder Hey Children's NHS Foundation Trust, Liverpool, United Kingdom
| | - Rajeev Shukla
- Department of Perinatal and Paediatric Pathology, Alder Hey Children's NHS Foundation Trust, Liverpool, United Kingdom
| | - Antonio Pizzuti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Anna Clerico
- Department of Paediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| | - Carlo Dominici
- Department of Paediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
26
|
Zhao W, Liu Z, Yu X, Lai L, Li H, Liu Z, Li L, Jiang S, Xia Z, Xu SY. iTRAQ proteomics analysis reveals that PI3K is highly associated with bupivacaine-induced neurotoxicity pathways. Proteomics 2016; 16:564-75. [PMID: 26621341 DOI: 10.1002/pmic.201500202] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 10/26/2015] [Accepted: 11/14/2015] [Indexed: 01/09/2023]
Abstract
Bupivacaine, a commonly used local anesthetic, has potential neurotoxicity through diverse signaling pathways. However, the key mechanism of bupivacaine-induced neurotoxicity remains unclear. Cultured human SH-SY5Y neuroblastoma cells were treated (bupivacaine) or untreated (control) with bupivacaine for 24 h. Compared to the control group, bupivacaine significantly increased cyto-inhibition, cellular reactive oxygen species, DNA damage, mitochondrial injury, apoptosis (increased TUNEL-positive cells, cleaved caspase 3, and Bcl-2/Bax), and activated autophagy (enhanced LC3II/LC3I ratio). To explore changes in protein expression and intercommunication among the pathways involved in bupivacaine-induced neurotoxicity, an 8-plex iTRAQ proteomic technique and bioinformatics analysis were performed. Compared to the control group, 241 differentially expressed proteins were identified, of which, 145 were up-regulated and 96 were down-regulated. Bioinformatics analysis of the cross-talk between the significant proteins with altered expression in bupivacaine-induced neurotoxicity indicated that phosphatidyl-3-kinase (PI3K) was the most frequently targeted protein in each of the interactions. We further confirmed these results by determining the downstream targets of the identified signaling pathways (PI3K, Akt, FoxO1, Erk, and JNK). In conclusion, our study demonstrated that PI3K may play a central role in contacting and regulating the signaling pathways that contribute to bupivacaine-induced neurotoxicity.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangdong Province, P. R. China
| | - Zhongjie Liu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangdong Province, P. R. China
| | - Xujiao Yu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangdong Province, P. R. China
| | - Luying Lai
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangdong Province, P. R. China
| | - Haobo Li
- Department of Anesthesiology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China.,Department of Anesthesiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, P. R. China
| | - Zipeng Liu
- Department of Anesthesiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, P. R. China
| | - Le Li
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangdong Province, P. R. China
| | - Shan Jiang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangdong Province, P. R. China
| | - Zhengyuan Xia
- Department of Anesthesiology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P. R. China.,Department of Anesthesiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, P. R. China
| | - Shi-yuan Xu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangdong Province, P. R. China
| |
Collapse
|
27
|
Maurya AK, Vinayak M. PI-103 and Quercetin Attenuate PI3K-AKT Signaling Pathway in T- Cell Lymphoma Exposed to Hydrogen Peroxide. PLoS One 2016; 11:e0160686. [PMID: 27494022 PMCID: PMC4975451 DOI: 10.1371/journal.pone.0160686] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 07/24/2016] [Indexed: 12/15/2022] Open
Abstract
Phosphatidylinositol 3 kinase-protein kinase B (PI3K-AKT) pathway has been considered as major drug target site due to its frequent activation in cancer. AKT regulates the activity of various targets to promote tumorigenesis and metastasis. Accumulation of reactive oxygen species (ROS) has been linked to oxidative stress and regulation of signaling pathways for metabolic adaptation of tumor microenvironment. Hydrogen peroxide (H2O2) in this context is used as ROS source for oxidative stress preconditioning. Antioxidants are commonly considered to be beneficial to reduce detrimental effects of ROS and are recommended as dietary supplements. Quercetin, a ubiquitous bioactive flavonoid is a dietary component which has attracted much of interest due to its potential health-promoting effects. Present study is aimed to analyze PI3K-AKT signaling pathway in H2O2 exposed Dalton's lymphoma ascite (DLA) cells. Further, regulation of PI3K-AKT pathway by quercetin as well as PI-103, an inhibitor of PI3K was analyzed. Exposure of H2O2 (1mM H2O2 for 30min) to DLA cells caused ROS accumulation and resulted in increased phosphorylation of PI3K and downstream proteins PDK1 and AKT (Ser-473 and Thr-308), cell survival factors BAD and ERK1/2, as well as TNFR1. However, level of tumor suppressor PTEN was declined. Both PI-103 & quercetin suppressed the enhanced level of ROS and significantly down-regulated phosphorylation of AKT, PDK1, BAD and level of TNFR1 as well as increased the level of PTEN in H2O2 induced lymphoma cells. The overall result suggests that quercetin and PI3K inhibitor PI-103 attenuate PI3K-AKT pathway in a similar mechanism.
Collapse
Affiliation(s)
- Akhilendra Kumar Maurya
- Biochemistry & Molecular Biology Laboratory, Centre for Advanced Study in Zoology, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - Manjula Vinayak
- Biochemistry & Molecular Biology Laboratory, Centre for Advanced Study in Zoology, Institute of Science, Banaras Hindu University, Varanasi-221005, India
- * E-mail:
| |
Collapse
|
28
|
Bell ES, Lammerding J. Causes and consequences of nuclear envelope alterations in tumour progression. Eur J Cell Biol 2016; 95:449-464. [PMID: 27397692 DOI: 10.1016/j.ejcb.2016.06.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 12/31/2022] Open
Abstract
Morphological changes in the size and shape of the nucleus are highly prevalent in cancer, but the underlying molecular mechanisms and the functional relevance remain poorly understood. Nuclear envelope proteins, which can modulate nuclear shape and organization, have emerged as key components in a variety of signalling pathways long implicated in tumourigenesis and metastasis. The expression of nuclear envelope proteins is altered in many cancers, and changes in levels of nuclear envelope proteins lamins A and C are associated with poor prognosis in multiple human cancers. In this review we highlight the role of the nuclear envelope in different processes important for tumour initiation and cancer progression, with a focus on lamins A and C. Lamin A/C controls many cellular processes with key roles in cancer, including cell invasion, stemness, genomic stability, signal transduction, transcriptional regulation, and resistance to mechanical stress. In addition, we discuss potential mechanisms mediating the changes in lamin levels observed in many cancers. A better understanding of cause-and-effect relationships between lamin expression and tumour progression could reveal important mechanisms for coordinated regulation of oncogenic processes, and indicate therapeutic vulnerabilities that could be exploited for improved patient outcome.
Collapse
Affiliation(s)
- Emily S Bell
- Meinig School of Biomedical Engineering & Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, United States
| | - Jan Lammerding
- Meinig School of Biomedical Engineering & Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, United States.
| |
Collapse
|
29
|
Ciccarelli C, Vulcano F, Milazzo L, Gravina GL, Marampon F, Macioce G, Giampaolo A, Tombolini V, Di Paolo V, Hassan HJ, Zani BM. Key role of MEK/ERK pathway in sustaining tumorigenicity and in vitro radioresistance of embryonal rhabdomyosarcoma stem-like cell population. Mol Cancer 2016; 15:16. [PMID: 26897742 PMCID: PMC4761200 DOI: 10.1186/s12943-016-0501-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 02/13/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The identification of signaling pathways that affect the cancer stem-like phenotype may provide insights into therapeutic targets for combating embryonal rhabdomyosarcoma. The aim of this study was to investigate the role of the MEK/ERK pathway in controlling the cancer stem-like phenotype using a model of rhabdospheres derived from the embryonal rhabdomyosarcoma cell line (RD). METHODS Rhabdospheres enriched in cancer stem like cells were obtained growing RD cells in non adherent condition in stem cell medium. Stem cell markers were evaluated by FACS analysis and immunoblotting. ERK1/2, myogenic markers, proteins of DNA repair and bone marrow X-linked kinase (BMX) expression were evaluated by immunoblotting analysis. Radiation was delivered using an x-6 MV photon linear accelerator. Xenografts were obtained in NOD/SCID mice by subcutaneously injection of rhabdosphere cells or cells pretreated with U0126 in stem cell medium. RESULTS MEK/ERK inhibitor U0126 dramatically prevented rhabdosphere formation and down-regulated stem cell markers CD133, CXCR4 and Nanog expression, but enhanced ALDH, MAPK phospho-active p38 and differentiative myogenic markers. By contrast, MAPK p38 inhibition accelerated rhabdosphere formation and enhanced phospho-active ERK1/2 and Nanog expression. RD cells, chronically treated with U0126 and then xeno-transplanted in NOD/SCID mice, delayed tumor development and reduced tumor mass when compared with tumor induced by rhabdosphere cells. U0126 intraperitoneal administration to mice bearing rhabdosphere-derived tumors inhibited tumor growth . The MEK/ERK pathway role in rhabdosphere radiosensitivity was investigated in vitro. Disassembly of rhabdospheres was induced by both radiation or U0126, and further enhanced by combined treatment. In U0126-treated rhabdospheres, the expression of the stem cell markers CD133 and CXCR4 decreased and dropped even more markedly following combined treatment. The expression of BMX, a negative regulator of apoptosis, also decreased following combined treatment, which suggests an increase in radiosensitivity of rhabdosphere cells. CONCLUSIONS Our results indicate that the MEK/ERK pathway plays a prominent role in maintaining the stem-like phenotype of RD cells, their survival and their innate radioresistance. Thus, therapeutic strategies that target cancer stem cells, which are resistant to traditional cancer therapies, may benefit from MEK/ERK inhibition combined with traditional radiotherapy, thereby providing a promising therapy for embryonal rhabdomyosarcoma.
Collapse
Affiliation(s)
- Carmela Ciccarelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy.
| | - Francesca Vulcano
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Luisa Milazzo
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy.
| | - Francesco Marampon
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy.
| | - Giampiero Macioce
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Adele Giampaolo
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | | | - Virginia Di Paolo
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy.
| | - Hamisa Jane Hassan
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Bianca Maria Zani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy.
| |
Collapse
|
30
|
Arderiu G, Espinosa S, Peña E, Aledo R, Badimon L. PAR2-SMAD3 in microvascular endothelial cells is indispensable for vascular stability via tissue factor signaling. J Mol Cell Biol 2015; 8:255-70. [PMID: 26658897 DOI: 10.1093/jmcb/mjv065] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/29/2015] [Indexed: 12/31/2022] Open
Abstract
Tissue factor (TF) signaling regulates gene expression and protein synthesis leading to the modulation of cell function. Recently, we have demonstrated in microvascular endothelial cells (mECs) that TF signaling induces activation of ETS1 transcription factor. Because combinatorial control is a characteristic property of ETS family members, involving the interaction between ETS1 and other transcription factors, here we investigate whether additional transcription factors are involved in TF-induced angiogenesis. We show by in vitro and in vivo experiments that in addition to ETS1, SMAD3 contributes to tube-like stabilization induced by TF in mECs. Whereas the ability of TF-overexpressing cells to induce gene expression through ETS1 is dependent on AKT signaling, SMAD3 induces ETS1 by an alternative AKT-independent pathway. Moreover, while TF-AKT-ETS1 pathway to induce CCL2 is PAR2-independent, PAR2 is required for TF-SMAD3-induced CCL2 expression. PAR2-dependent activation of SMAD3 is mediated by PKC phosphorylation. In addition, disruption of SMAD3 expression in mECs reduces ERK1/2 phosphorylation and decreases target gene promoter activity. In conclusion, in mECs TF-induced angiogenesis seems to be the result of two signaling pathways: TF-induced microvessel formation is regulated through β1 integrin-AKT-ETS1; and TF-induced microvessel stabilization is regulated via PAR2-SMAD3 that is indispensable for the maintenance of vascular integrity.
Collapse
Affiliation(s)
- Gemma Arderiu
- Cardiovascular Research Center (CSIC-ICCC), Hospital de Sant Pau (UAB) and IIB-Sant Pau, 08025 Barcelona, Spain
| | - Sonia Espinosa
- Cardiovascular Research Center (CSIC-ICCC), Hospital de Sant Pau (UAB) and IIB-Sant Pau, 08025 Barcelona, Spain
| | - Esther Peña
- Cardiovascular Research Center (CSIC-ICCC), Hospital de Sant Pau (UAB) and IIB-Sant Pau, 08025 Barcelona, Spain
| | - Rosa Aledo
- Cardiovascular Research Center (CSIC-ICCC), Hospital de Sant Pau (UAB) and IIB-Sant Pau, 08025 Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Research Center (CSIC-ICCC), Hospital de Sant Pau (UAB) and IIB-Sant Pau, 08025 Barcelona, Spain
| |
Collapse
|
31
|
Sun M, Huang F, Yu D, Zhang Y, Xu H, Zhang L, Li L, Dong L, Guo L, Wang S. Autoregulatory loop between TGF-β1/miR-411-5p/SPRY4 and MAPK pathway in rhabdomyosarcoma modulates proliferation and differentiation. Cell Death Dis 2015; 6:e1859. [PMID: 26291313 PMCID: PMC4558514 DOI: 10.1038/cddis.2015.225] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 05/13/2015] [Accepted: 07/07/2015] [Indexed: 12/30/2022]
Abstract
The origin of rhabdomyosarcoma (RMS) remains controversial. However, specific microRNAs (miRNAs) are downregulated in RMS and it is possible that re-expression of these miRNAs may lead to differentiation. Transforming growth factor-β1 (TGF-β1) is known to block differentiation of RMS. We therefore analyzed miRNA microarrays of RMS cell lines with or without TGF-β1 knockdown and identified a novel anti-oncogene miR-411-5p. Re-expression of miR-411-5p inhibited RMS cell proliferation in vitro and tumorigenicity in vivo. Using a luciferase reporting system and sequence analysis, the potential target of miR-411-5p was identified as sprouty homolog 4 (SPRY4), which inhibits protein kinase Cα-mediated activation of mitogen-activated protein kinases (MAPKs), especially p38MAPK phosphorylation. These results revealed an inverse correlation between TGF-β1/SPRY4 and miR-411-5p levels. SPRY4 small interfering RNA and miR-411-5p both activated p38MAPK phosphorylation and also promoted apoptosis and myogenic differentiation, indicated by increased caspase-3, myosin heavy chain, and myosin expression. SPRY4 and miR-411 mRNA levels correlated with TGF-β1 expression levels in RMS tissues, which was confirmed by immunohistochemical staining for TGF-β1, SPRY4, and phosphorylated p38MAPK proteins. Overall, these results indicate that miR-411-5p acts as an RMS differentiation-inducing miRNA prompting p38MAPK activation via directly downregulating SPRY4. These results establish an autoregulatory loop between TGF-β1/miR-411-5p/SPRY4 and MAPK in RMS, which governs the switch between proliferation and differentiation.
Collapse
Affiliation(s)
- M Sun
- Laboratory Animal Research Center, Soochow University School of Medicine, Suzhou 215123, China
| | - F Huang
- Department of Pathology, Soochow University School of Medicine, Suzhou 215123, China
| | - D Yu
- Department of Plastic Surgery, Second Affiliated Hospital, Soochow University, Suzhou 215004, China
| | - Y Zhang
- Department of Pathology, Soochow University School of Medicine, Suzhou 215123, China
| | - H Xu
- Department of Oncology, First Affiliated Hospital, Soochow University, Suzhou 215006, China
| | - L Zhang
- Department of Surgery, First Affiliated Hospital, Soochow University, Suzhou 215006, China
| | - L Li
- Department of Osteology, Second Affiliated Hospital, Soochow University, Suzhou 215004, China
| | - L Dong
- Department of Pathology, Soochow University School of Medicine, Suzhou 215123, China
| | - L Guo
- Department of Pathology, Soochow University School of Medicine, Suzhou 215123, China
| | - S Wang
- Department of Pathology, Soochow University School of Medicine, Suzhou 215123, China
| |
Collapse
|
32
|
Maurya AK, Vinayak M. Modulation of PKC signaling and induction of apoptosis through suppression of reactive oxygen species and tumor necrosis factor receptor 1 (TNFR1): key role of quercetin in cancer prevention. Tumour Biol 2015; 36:8913-24. [DOI: 10.1007/s13277-015-3634-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/01/2015] [Indexed: 02/06/2023] Open
|
33
|
Walker N, Kahamba T, Woudberg N, Goetsch K, Niesler C. Dose-dependent modulation of myogenesis by HGF: implications for c-Met expression and downstream signalling pathways. Growth Factors 2015; 33:229-41. [PMID: 26135603 DOI: 10.3109/08977194.2015.1058260] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Hepatocyte growth factor (HGF) regulates satellite cell activation, proliferation, and differentiation. We analyzed the dose-dependent effects of HGF on myogenesis. Murine C2C12 and human donor-derived skeletal muscle myoblasts were treated with 0, 2, or 10 ng/ml HGF followed by assessment of proliferation and differentiation. HGF (2 ng/ml) significantly promoted cell division, but reduced myogenic commitment and fusion. Conversely, 10 ng/ml HGF reduced proliferative capability, but increased differentiation. c-Met expression analysis revealed significantly decreased expression in differentiating cells cultured with 2 ng/ml HGF, but increased expression in proliferating cells with 10 ng/ml HGF. Mitogen-activated protein kinase (MAPKs: ERK, JNK, or p38K) and phosphatidylinositol-3-kinase (PI3K) inhibition abrogated the HGF-stimulated increase in cell number. Interestingly, PI3K and p38 kinase facilitated the negative effect of HGF on proliferation, while ERK inhibition abrogated the HGF-mediated decrease in differentiation. Dose-dependent effects of HGF are mediated by changes in c-Met expression and downstream MAPK and PI3K signalling.
Collapse
Affiliation(s)
- Nicholas Walker
- a Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal , Scottsville , South Africa
| | - Trish Kahamba
- a Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal , Scottsville , South Africa
| | - Nicholas Woudberg
- a Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal , Scottsville , South Africa
| | - Kyle Goetsch
- a Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal , Scottsville , South Africa
| | - Carola Niesler
- a Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal , Scottsville , South Africa
| |
Collapse
|
34
|
HUANG RONGRONG, HU WEN, YIN YANYAN, WANG YUCHAN, LI WEIPING, LI WEIZU. Chronic restraint stress promotes learning and memory impairment due to enhanced neuronal endoplasmic reticulum stress in the frontal cortex and hippocampus in male mice. Int J Mol Med 2014; 35:553-9. [DOI: 10.3892/ijmm.2014.2026] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 12/01/2014] [Indexed: 11/06/2022] Open
|
35
|
Zhang P, Goodrich C, Fu C, Dong C. Melanoma upregulates ICAM-1 expression on endothelial cells through engagement of tumor CD44 with endothelial E-selectin and activation of a PKCα-p38-SP-1 pathway. FASEB J 2014; 28:4591-609. [PMID: 25138157 DOI: 10.1096/fj.11-202747] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer metastasis involves multistep adhesive interactions between tumor cells (TCs) and endothelial cells (ECs), but the molecular mechanisms of intercellular communication in the tumor microenvironment remain elusive. Using static and flow coculture systems in conjunction with flow cytometry, we discovered that certain receptors on the ECs are upregulated on melanoma cell adhesion. Direct contact but not separate coculture between human umbilical endothelial cells (HUVECs) and a human melanoma cell line (Lu1205) increased intercellular adhesion molecule 1 (ICAM-1) and E-selectin expression on HUVECs by 3- and 1.5-fold, respectively, compared with HUVECs alone. The nonmetastatic cell line WM35 failed to promote ICAM-1 expression changes in HUVECs on contact. Enzyme-linked immunosorbent assay (ELISA) revealed that EC-TC contact has a synergistic effect on the expression of the cytokines interleukin (IL)-8, IL-6, and growth-related oncogene α (Gro-α). By using E-selectin cross-linking and beads coated with CD44 immunopurified from Lu1205 cells, we showed that CD44/selectin ligation was responsible for the ICAM-1 up-regulation on HUVECs. Protein kinase Cα (PKC-α) activation was found to be the downstream target of the CD44/selectin-initiated signaling, as ICAM-1 elevation was inhibited by siRNA targeting PKCα or a dominant negative form of PKCα (PKCα DN). Western blot analysis and electrophoretic mobility shift assays (EMSAs) showed that TC-EC contact mediated p38 phosphorylation and binding of the transcription factor SP-1 to its regulation site. In conclusion, CD44/selectin binding signals ICAM-1 up-regulation on the EC surface through a PKCα-p38-SP-1 pathway, which further enhances melanoma cell adhesion to ECs during metastasis.
Collapse
Affiliation(s)
- Pu Zhang
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Chris Goodrich
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Changliang Fu
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Cheng Dong
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
36
|
Wu G, Song C, Lu H, Jia L, Yang G, Shi X, Sun S. Sirt2 induces C2C12 myoblasts proliferation by activation of the ERK1/2 pathway. Acta Biochim Biophys Sin (Shanghai) 2014; 46:342-5. [PMID: 24457518 DOI: 10.1093/abbs/gmt151] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Guofang Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | | | | | | | | | | | | |
Collapse
|
37
|
Carduner L, Picot CR, Leroy-Dudal J, Blay L, Kellouche S, Carreiras F. Cell cycle arrest or survival signaling through αv integrins, activation of PKC and ERK1/2 lead to anoikis resistance of ovarian cancer spheroids. Exp Cell Res 2013; 320:329-42. [PMID: 24291221 DOI: 10.1016/j.yexcr.2013.11.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 11/11/2013] [Accepted: 11/14/2013] [Indexed: 11/24/2022]
Abstract
Ovarian cancer is the most lethal gynecologic cancer mainly due to spheroids organization of cancer cells that disseminate within the peritoneal cavity. We have investigated the molecular mechanisms by which ovarian cancer spheroids resist anoikis, choosing as models the 2 well-characterized human ovarian cancer cell lines IGROV1 and SKOV3. These cell lines have the propensity to float as clusters, and were isolated from tumor tissue and ascites, respectively. To form spheroids, IGROV1 and SKOV3 ovarian adenocarcinoma cells were maintained under anchorage-independent culture conditions, in which both lines survive at least a week. A short apoptotic period prior to a survival signaling commitment was observed for IGROV1 cells whereas SKOV3 cells entered G0/G1 phase of the cell cycle. This difference in behavior was due to different signals. With regard to SKOV3 cells, activation of p38 and an increase in p130/Rb occurred once anchorage-independent culture was established. Analyses of the survival signaling pathway switched on by IGROV1 cells showed that activation of ERK1/2 was required to evade apoptosis, an effect partly dependent on PKC activation and αv integrins. αv-integrin expression is essential for survival through activation of ERK1/2 phosphorylation. The above data indicate that ovarian cancer cells can resist anoikis in the spheroid state by arrest in the cell cycle or through activation of αv-integrin-ERK-mediated survival signals. Such signaling might result in the selection of resistant cells within disseminating spheroids, favoring further relapse in ovarian cancers.
Collapse
Affiliation(s)
- Ludovic Carduner
- Equipe de Recherche sur les Relations Matrice Extracellulaire Cellules, ERRMECe (EA 1391), Institut des Matériaux, Université de Cergy-Pontoise, 2 avenue Adolphe Chauvin, 95302 Cergy-Pontoise Cedex, France
| | - Cédric R Picot
- Equipe de Recherche sur les Relations Matrice Extracellulaire Cellules, ERRMECe (EA 1391), Institut des Matériaux, Université de Cergy-Pontoise, 2 avenue Adolphe Chauvin, 95302 Cergy-Pontoise Cedex, France.
| | - Johanne Leroy-Dudal
- Equipe de Recherche sur les Relations Matrice Extracellulaire Cellules, ERRMECe (EA 1391), Institut des Matériaux, Université de Cergy-Pontoise, 2 avenue Adolphe Chauvin, 95302 Cergy-Pontoise Cedex, France
| | - Lyvia Blay
- Equipe de Recherche sur les Relations Matrice Extracellulaire Cellules, ERRMECe (EA 1391), Institut des Matériaux, Université de Cergy-Pontoise, 2 avenue Adolphe Chauvin, 95302 Cergy-Pontoise Cedex, France
| | - Sabrina Kellouche
- Equipe de Recherche sur les Relations Matrice Extracellulaire Cellules, ERRMECe (EA 1391), Institut des Matériaux, Université de Cergy-Pontoise, 2 avenue Adolphe Chauvin, 95302 Cergy-Pontoise Cedex, France
| | - Franck Carreiras
- Equipe de Recherche sur les Relations Matrice Extracellulaire Cellules, ERRMECe (EA 1391), Institut des Matériaux, Université de Cergy-Pontoise, 2 avenue Adolphe Chauvin, 95302 Cergy-Pontoise Cedex, France
| |
Collapse
|
38
|
Zucchini-Pascal N, Peyre L, Rahmani R. Crosstalk between beta-catenin and snail in the induction of epithelial to mesenchymal transition in hepatocarcinoma: role of the ERK1/2 pathway. Int J Mol Sci 2013; 14:20768-92. [PMID: 24135872 PMCID: PMC3821642 DOI: 10.3390/ijms141020768] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 09/23/2013] [Accepted: 10/03/2013] [Indexed: 12/14/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is an integral process in the progression of many epithelial tumors. It involves a coordinated series of events, leading to the loss of epithelial features and the acquisition of a mesenchymal phenotype, resulting in invasion and metastasis. The EMT of hepatocellular carcinoma (HCC) cells is thought to be a key event in intrahepatic dissemination and distal metastasis. In this study, we used 12-O-tet-radecanoylphorbol-13-acetate (TPA) to dissect the signaling pathways involved in the EMT of HepG2 hepatocarcinoma cells. The spectacular change in phenotype induced by TPA, leading to a pronounced spindle-shaped fibroblast-like cell morphology, required ERK1/2 activation. This ERK1/2-dependent EMT process was characterized by a loss of E-cadherin function, modification of the cytoskeleton, the acquisition of mesenchymal markers and profound changes to extracellular matrix composition and mobility. Snail was essential for E-cadherin repression, but was not sufficient for full commitment of the TPA-triggered EMT. We found that TPA triggered the formation of a complex between Snail and β-catenin that activated the Wnt pathway. This study thus provides the first evidence for the existence of a complex network governed by the ERK1/2 signaling pathway, converging on the coregulation of Snail and the Wnt/β-catenin pathway and responsible for the onset and the progression of EMT in hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Nathalie Zucchini-Pascal
- Laboratory of Xenobiotic's Cellular and Molecular Toxicology, INRA, UMR 1331 TOXALIM (Research Centre in Food Toxicology), Sophia Antipolis 06903, France.
| | | | | |
Collapse
|
39
|
Keller C, Guttridge DC. Mechanisms of impaired differentiation in rhabdomyosarcoma. FEBS J 2013; 280:4323-34. [PMID: 23822136 DOI: 10.1111/febs.12421] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 06/17/2013] [Accepted: 07/01/2013] [Indexed: 12/22/2022]
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood, with presumed skeletal muscle origins, because of its myogenic phenotype. RMS is composed of two main subtypes, embryonal RMS (eRMS) and alveolar RMS (aRMS). Whereas eRMS histologically resembles embryonic skeletal muscle, the aRMS subtype is more aggressive and has a poorer prognosis. In addition, whereas the genetic profile of eRMS is not well established, aRMS is commonly associated with distinct chromosome translocations that fuse domains of the transcription factors Pax3 and Pax7 to the forkhead family member FOXO1A. Both eRMS and aRMS tumor cells express myogenic markers such as MyoD, but their ability to complete differentiation is impaired. How this impairment occurs is the subject of this review, which will focus on several themes, including signaling pathways that converge on Pax-forkhead gene targets, alterations in MyoD function, epigenetic modifications of myogenic promoters, and microRNAs whose expression patterns in RMS alter key regulatory circuits to help maintain tumor cells in an opportunistically less differentiated state.
Collapse
Affiliation(s)
- Charles Keller
- Pediatric Cancer Biology Program, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | | |
Collapse
|
40
|
Ghorpade DS, Holla S, Sinha AY, Alagesan SK, Balaji KN. Nitric oxide and KLF4 protein epigenetically modify class II transactivator to repress major histocompatibility complex II expression during Mycobacterium bovis bacillus Calmette-Guerin infection. J Biol Chem 2013; 288:20592-606. [PMID: 23733190 DOI: 10.1074/jbc.m113.472183] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pathogenic mycobacteria employ several immune evasion strategies such as inhibition of class II transactivator (CIITA) and MHC-II expression, to survive and persist in host macrophages. However, precise roles for specific signaling components executing down-regulation of CIITA/MHC-II have not been adequately addressed. Here, we demonstrate that Mycobacterium bovis bacillus Calmette-Guérin (BCG)-mediated TLR2 signaling-induced iNOS/NO expression is obligatory for the suppression of IFN-γ-induced CIITA/MHC-II functions. Significantly, NOTCH/PKC/MAPK-triggered signaling cross-talk was found critical for iNOS/NO production. NO responsive recruitment of a bifunctional transcription factor, KLF4, to the promoter of CIITA during M. bovis BCG infection of macrophages was essential to orchestrate the epigenetic modifications mediated by histone methyltransferase EZH2 or miR-150 and thus calibrate CIITA/MHC-II expression. NO-dependent KLF4 regulated the processing and presentation of ovalbumin by infected macrophages to reactive T cells. Altogether, our study delineates a novel role for iNOS/NO/KLF4 in dictating the mycobacterial capacity to inhibit CIITA/MHC-II-mediated antigen presentation by infected macrophages and thereby elude immune surveillance.
Collapse
Affiliation(s)
- Devram Sampat Ghorpade
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | | | | | | | | |
Collapse
|
41
|
Stoppani E, Rossi S, Marchesini S, Preti A, Fanzani A. Defective myogenic differentiation of human rhabdomyosarcoma cells is characterized by sialidase Neu2 loss of expression. Cell Biol Int 2013; 33:1020-5. [DOI: 10.1016/j.cellbi.2009.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 04/30/2009] [Accepted: 06/03/2009] [Indexed: 11/16/2022]
|
42
|
Yang H, Li F, Kong X, Yuan X, Wang W, Huang R, Li T, Geng M, Wu G, Yin Y. Chemerin regulates proliferation and differentiation of myoblast cells via ERK1/2 and mTOR signaling pathways. Cytokine 2012; 60:646-52. [DOI: 10.1016/j.cyto.2012.07.033] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 07/13/2012] [Accepted: 07/29/2012] [Indexed: 10/28/2022]
|
43
|
Hepatitis C virus activates Bcl-2 and MMP-2 expression through multiple cellular signaling pathways. J Virol 2012; 86:12531-43. [PMID: 22951829 DOI: 10.1128/jvi.01136-12] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hepatitis C virus (HCV) infection is associated with numerous liver diseases and causes serious global health problems, but the mechanisms underlying the pathogenesis of HCV infections remain largely unknown. In this study, we demonstrate that signal transducer and activator of transcription 3 (STAT3), matrix metalloproteinase-2 (MMP-2), and B-cell lymphoma 2 (Bcl-2) are significantly stimulated in HCV-infected patients. We further show that HCV activates STAT3, MMP-2, Bcl-2, extracellular regulated protein kinase (ERK), and c-Jun N-terminal kinase (JNK) in infected Huh7.5.1 cells. Functional screening of HCV proteins revealed that nonstructural protein 4B (NS4B) is responsible for the activation of MMP-2 and Bcl-2 by stimulating STAT3 through repression of the suppressor of cytokine signaling 3 (SOCS3). Our results also demonstrate that multiple signaling cascades, including several members of the protein kinase C (PKC) family, JNK, ERK, and STAT3, play critical roles in the activation of MMP-2 and Bcl-2 mediated by NS4B. Further studies revealed that the C-terminal domain (CTD) of NS4B is sufficient for the activation of STAT3, JNK, ERK, MMP-2, and Bcl-2. We also show that amino acids 227 to 250 of NS4B are essential for regulation of STAT3, JNK, ERK, MMP-2, and Bcl-2, and among them, three residues (237L, 239S, and 245L) are crucial for this regulation. Thus, we reveal a novel mechanism underlying HCV pathogenesis in which multiple intracellular signaling cascades are cooperatively involved in the activation of two important cellular factors, MMP-2 and Bcl-2, in response to HCV infection.
Collapse
|
44
|
Foster JD, Yang JW, Moritz AE, Challasivakanaka S, Smith MA, Holy M, Wilebski K, Sitte HH, Vaughan RA. Dopamine transporter phosphorylation site threonine 53 regulates substrate reuptake and amphetamine-stimulated efflux. J Biol Chem 2012; 287:29702-12. [PMID: 22722938 PMCID: PMC3436161 DOI: 10.1074/jbc.m112.367706] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
In the central nervous system, levels of extraneuronal dopamine are controlled primarily by the action of the dopamine transporter (DAT). Multiple signaling pathways regulate transport activity, substrate efflux, and other DAT functions through currently unknown mechanisms. DAT is phosphorylated by protein kinase C within a serine cluster at the distal end of the cytoplasmic N terminus, whereas recent work in model cells revealed proline-directed phosphorylation of rat DAT at membrane-proximal residue Thr(53). In this report, we use mass spectrometry and a newly developed phospho-specific antibody to positively identify DAT phosphorylation at Thr(53) in rodent striatal tissue and heterologous expression systems. Basal phosphorylation of Thr(53) occurred with a stoichiometry of ~50% and was strongly increased by phorbol esters and protein phosphatase inhibitors, demonstrating modulation of the site by signaling pathways that impact DAT activity. Mutations of Thr(53) to prevent phosphorylation led to reduced dopamine transport V(max) and total apparent loss of amphetamine-stimulated substrate efflux, supporting a major role for this residue in the transport kinetic mechanism.
Collapse
Affiliation(s)
- James D Foster
- Department of Biochemistry and Molecular Biology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202-9037, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Lin BR, Natarajan V. Negative regulation of human U6 snRNA promoter by p38 kinase through Oct-1. Gene 2012; 497:200-7. [PMID: 22310390 PMCID: PMC3306512 DOI: 10.1016/j.gene.2012.01.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 01/06/2012] [Accepted: 01/18/2012] [Indexed: 01/08/2023]
Abstract
Recruitment of Oct-1 protein to the octamer sequence of U6 promoter is critical for optimal transcription by RNA polymerase III. Here we report that p38 kinase inhibitors, SB202190 and SB203580, stimulated U6 promoter activity and this stimulation can be observed only in the presence of octamer sequence. SB202190-treated cell nuclear extract had about 50% increase in Oct-1 binding activity suggesting that the increased U6 promoter activity by p38 kinase inhibitor is mediated through Oct-1. Mutation in octamer sequence significantly reduced the SB202190-stimulated U6 promoter transcription and the distance between octamer and proximal sequence element of U6 promoter is also critical for the p38 kinase inhibitor-stimulated activity. Exogenous Oct-1 expression showed a concentration-dependent activation of U6 promoter that was further stimulated by the p38 kinase inhibitors. When cells were treated with p38 kinase inducer, hydrogen peroxide or phorbol 12-myristate 13-acetate (PMA), U6 promoter activity was down regulated and this inhibition was reversed by p38 kinase inhibitors. Over-expression of p38α kinase down-regulated U6 promoter activity and this inhibition was further enhanced by PMA and p38 kinase inhibitors reversed this inhibition. p38 kinase inhibitor-treated cells had 50% more U6 RNA than the control cells. Taken together, our results show a negative correlation between the p38 kinase levels and Oct-1 binding on U6 promoter, suggesting that U6 promoter is negatively regulated by p38 kinase.
Collapse
Affiliation(s)
- Bor-Ruei Lin
- SAIC-Frederick, National Cancer Institute at Frederick, 1050 Boyles Street, Frederick, MD 21702-1201, USA
| | - Ven Natarajan
- SAIC-Frederick, National Cancer Institute at Frederick, 1050 Boyles Street, Frederick, MD 21702-1201, USA
| |
Collapse
|
46
|
Velpula KK, Dasari VR, Tsung AJ, Dinh DH, Rao JS. Transcriptional repression of Mad-Max complex by human umbilical cord blood stem cells downregulates extracellular signal-regulated kinase in glioblastoma. Stem Cells Dev 2011; 21:1779-93. [PMID: 21933022 DOI: 10.1089/scd.2011.0424] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Previously, we have shown that human umbilical cord blood stem cell (hUCBSC) treatment downregulate cyclin D1 in glioma cells. To study the cell cycle progression and investigate the upstream molecules regulating cyclin D1 expression, we analyzed the involvement of extracellular signal-regulated kinase (ERK) and its functionality after treatment with hUCBSC. We observed downregulation of pERK after hUCBSC treatment at both transcriptional and translational levels. Increased translocation of ERK from cytoplasm to the nucleus was observed in glioma cells, whereas hUCBSC cocultures with glioma cells showed suppressed nuclear translocation. This finding suggests that hUCBSC regulates ERK by suppressing its phosphorylation at phospho-Thr(202)/Tyr(204) retarding pERK nuclear translocation. ERK promoter analysis has shown c-Myc binding sites, indicative of possible transcriptional interactions that regulate cyclin D1 and ERK expression levels. Treatment of U251 and 5310 glioma cells with U0126, a MEK/ERK inhibitor receded pERK and c-Myc levels. In another experiment, U251 and 5310 cells treated with 10074-G5, c-Myc/Max inhibitor displayed reduction in pERK and c-Myc levels suggestive of a positive feedback loop between ERK/c-Myc/Max molecules. In the present study, we show that glioma cells exhibit abundant c-Myc expression and increased c-Myc/Max activity. In contrast, the glioma cells cocultured with hUCBSC demonstrated high Mad1 expression that competitively binds to Max to repress the c-Myc/Max mediated gene transcription. Our studies thus elucidate the potential role of hUCBSC in controlling glioma cell cycle progression and invasion by limiting Max binding to c-Myc, thus regulating the expression of glioma cell cycle and invasion associated molecules such as ERK, integrins via increased levels of Mad1 expression.
Collapse
Affiliation(s)
- Kiran Kumar Velpula
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL 61656, USA
| | | | | | | | | |
Collapse
|
47
|
Low-molecular-weight fucoidan regulates myogenic differentiation through the mitogen-activated protein kinase pathway in C2C12 cells. Br J Nutr 2011; 106:1836-44. [PMID: 21682942 DOI: 10.1017/s0007114511002534] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Low-molecular-weight fucoidan (LMWF) has been broadly studied in recent years due to its numerous biological properties. Nevertheless, there have been no reports about the effects of LMWF on myogenic differentiation (MyoD). The objective of the present study was to demonstrate the impact of LMWF on myogenesis in C2C12 cells. The ultimate aim was to establish whether LMWF regulates myogenesis similar to heparin as a partial regulator of myogenesis. LMWF was prepared at a minimal size by ultra-filtration compared with crude fucoidan. We treated C2C12 cells with LMWF and/or heparin during MyoD. The data from the present study are the first to suggest that LMWF suppresses the expression of the myogenic regulatory factors and the myocyte enhancer factors as well as the morphological changes that occur during differentiation. Additionally, the expression of the mitogen-activated protein kinase (MAPK) family was significantly inhibited by LMWF when compared with controls. The LMWF-treated group showed significantly decreased expression of reactive oxygen species (ROS) enzymes compared with control cells. Heparin was used as a positive control because it has been reported to activate MyoD. Taken together, these results suggest that LMWF might regulate MyoD through the MAPK pathway and by regulating ROS activity in C2C12 cells.
Collapse
|
48
|
Nakura A, Higuchi C, Yoshida K, Yoshikawa H. PKCα suppresses osteoblastic differentiation. Bone 2011; 48:476-84. [PMID: 20951242 DOI: 10.1016/j.bone.2010.09.238] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Revised: 09/08/2010] [Accepted: 09/29/2010] [Indexed: 11/30/2022]
Abstract
Protein kinase C (PKC) plays an essential role in cellular signal transduction for mediating a variety of biological functions. There are 11 PKC isoforms and these isoforms are believed to play distinct roles in cells. Although the role of individual isoforms of PKC has been investigated in many fields, little is known about the role of PKC in osteoblastic differentiation. Here, we investigated which isoforms of PKC are involved in osteoblastic differentiation of the mouse preosteoblastic cell line MC3T3-E1. Treatment with Gö6976, an inhibitor of PKCα and PKCβI, increased alkaline phosphatase (ALP) activity as well as gene expression of ALP and Osteocalcin (OCN), and enhanced calcification of the extracellular matrix. Concurrently, osteoblastic cell proliferation decreased at a concentration of 1.0 μM. In contrast, a PKCβ inhibitor, which inhibits PKCβI and PKCβII, did not significantly affect osteoblastic differentiation or cell proliferation. Knockdown of PKCα using MC3T3-E1 cells transfected with siRNA also induced an increase in ALP activity and in gene expression of ALP and OCN. In contrast, overexpression of wild-type PKCα decreased ALP activity and attenuated osteoblastic differentiation markers including ALP and OCN, but promoted cell proliferation. Taken together, our results indicate that PKCα suppresses osteoblastic differentiation, but promotes osteoblastic cell proliferation. These results imply that PKCα may have a pivotal role in cell signaling that modulates the differentiation and proliferation of osteoblasts.
Collapse
Affiliation(s)
- Akio Nakura
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | | | | | | |
Collapse
|
49
|
Rossi S, Stoppani E, Puri PL, Fanzani A. Differentiation of human rhabdomyosarcoma RD cells is regulated by reciprocal, functional interactions between myostatin, p38 and extracellular regulated kinase signalling pathways. Eur J Cancer 2011; 47:1095-105. [PMID: 21273059 DOI: 10.1016/j.ejca.2010.12.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 12/03/2010] [Accepted: 12/14/2010] [Indexed: 01/02/2023]
Abstract
Rhabdomyosarcoma (RMS) includes heterogeneous tumours of mesenchymal derivation which are genetically committed to the myogenic lineage, but fail to complete terminal differentiation. Previous works have reported on deregulated myostatin, p38 and extracellular regulated kinase (ERK) signalling in RMS cell lines; however, the functional link between these pathways and their relative contribution to RMS pathogenesis and/or maintenance of the transformed phenotype in vitro are unclear. Herein we show that the constitutive expression of a dominant-negative form of activin receptor type IIb (dnACTRIIb), which inhibits myostatin signalling, decreased proliferation and promoted differentiation of the human RMS RD cell line. DnACTRIIb-dependent differentiation of RD cells correlated with a reduced SMAD2/3 (small mother against decapentaplegic) and ERK signalling and the activation of p38 pathway. Conversely, the expression of a constitutively activated ALK5 (activin receptor-like kinase) (caALK5) form, activating SMAD3 and ERK pathways, led to further impairment of RD differentiation. Pharmacological blockade of ERK pathway in RD cells was sufficient to replicate the biological phenotype observed in dnACTRIIb-expressing RD cells, and also recovered the differentiation of caALK5-expressing RD cells. Conversely, deliberate activation of p38 signalling mimics the effect of dnActRIIb and overcame the differentiation block in RD cells. These data indicate the existence of a network formed by myostatin/SMAD2/3, ERK and p38 pathways that, when deregulated, might contribute to the pathogenesis of RMS. The components of this network might, therefore, be a valuable target for interventions towards correcting the malignant phenotype of RMS.
Collapse
Affiliation(s)
- Stefania Rossi
- Department of Biomedical Sciences and Biotechnologies and Interuniversity Institute of Myology (IIM), University of Brescia, viale Europa 11, 25123 Brescia, Italy
| | | | | | | |
Collapse
|
50
|
WJ9708012 exerts anticancer activity through PKC-α related crosstalk of mitochondrial and endoplasmic reticulum stresses in human hormone-refractory prostate cancer cells. Acta Pharmacol Sin 2011; 32:89-98. [PMID: 21132000 DOI: 10.1038/aps.2010.173] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AIM To investigate the anticancer mechanism of a methoxyflavanone derivative, WJ9708012, highlighting its role on a crosstalk between endoplasmic reticulum (ER) and mitochondrial stress. METHODS Cell proliferation was examined using sulforhodamine B assay. Cell-cycle progression, Ca(2+) mobilization and mitochondrial membrane potential (ΔΨ(m)) were detected using flow cytometric analysis. Protein expression was detected using Western blot. RESULTS WJ9708012 displayed an antiproliferative and apoptotic activity in human hormone-refractory prostate cancer cells with IC(50) values of 6.4 and 5.3 μmol/L in PC-3 and DU-145 cells. WJ9708012 induced a prompt increase of cytosolic Ca(2+) level and activation of protein kinase C (PKC)-α. The cleavage of μ-calpain was also induced by WJ9708012. Furthermore, WJ9708012 induced cell-cycle arrest at G(1)-phase associated with down-regulation of cyclin D1, cyclin E and cyclin-dependent kinase-4 expressions. It also caused a rapid and time-dependent decrease of phosphorylation level of mTOR (Ser(2448)), 4E-BP1 (Thr(37)/Thr(46)/Thr(70)) and p70S6K (Thr(389)), indicating the inhibition of mTOR-mediated translational pathways. The ER stress was activated by the identification of up-regulated GADD153 and glucose-regulated protein-78 protein levels. The subsequent mitochondrial stress was also identified by the observation of a decreased Bcl-2 and Bcl-xL expressions, an increased truncated Bid and Bad and a loss of ΔΨ(m). CONCLUSION WJ9708012 induces an increase of cytosolic Ca(2+) concentration and activation of PKC-α. Subsequently, a crosstalk between ER stress and mitochondrial insult is induced, leading to the inhibition of mTOR pathways and arrest of the cell-cycle at G(1) phase. The apoptosis is ultimately induced by a severe damage of mitochondrial function.
Collapse
|