Vol. 25 (2020)



No. 1DOI: 10.1186/s11658-019-0193-6 Volume 25 (2020) - 25:1
Title THE UBIQUITIN SYSTEM: ORCHESTRATING CELLULAR SIGNALS IN NON-SMALL-CELL LUNG CANCER
Authors Qiang Fan1,2†, Qian Wang1†, Renjie Cai, Haihua Yuan1* and Ming Xu1*
Abstract The ubiquitin system, known as a common feature in eukaryotes, participates in multiple cellular processes, such as signal transduction, cell-cycle progression, receptor trafficking and endocytosis, and even the immune response. In lung cancer, evidence has revealed that aberrant events in ubiquitin-mediated processes can cause a variety of pathological outcomes including tumorigenesis and metastasis. Likewise, ubiquitination on the core components contributing to the activity of cell signaling controls bio-signal turnover and cell final destination. Given this, inhibitors targeting the ubiquitin system have been developed for lung cancer therapies and have shown great prospects for clinical application. However, the exact biological effects and physiological role of the drugs used in lung cancer therapies are still not clearly elucidated, which might seriously impede the progress of treatment. In this work, we summarize current research advances in cell signal regulation processes mediated through the ubiquitin system during the development of lung cancer, with the hope of improving the therapeutic effects by means of aiming at efficient targets.
Keywords Ubiquitin, Ubiquitination, Deubiquitination, Cell signaling, Lung cancer
Address and Contact Information 1 Department of Oncology, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China
2 Department of General Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China.
* Corresponding author: ayuan790415@shsmu.edu.cn; mingxu.msu@gmail.com
Qiang Fan and Qian Wang contributed equally to this work.
Read full article at BMC

No. 2DOI: 10.1186/s11658-020-0199-0 Volume 25 (2020) - 25:2
Title DNA–DEPENDENT PROTEIN KINASE IN TELOMERE MAINTENANCE AND PROTECTION
Authors Jiangdong Sui1* , Shichuan Zhang2 and Benjamin P. C. Chen3*
Abstract This review focuses on DNA–dependent protein kinase (DNA–PK), which is the key regulator of canonical non–homologous end–joining (NHEJ), the predominant mechanism of DNA double–strand break (DSB) repair in mammals. DNA–PK consists of the DNA–binding Ku70/80 heterodimer and the catalytic subunit DNA–PKcs. They assemble at DNA ends, forming the active DNA–PK complex, which initiates NHEJ–mediated DSB repair. Paradoxically, both Ku and DNA–PKcs are associated with telomeres, and they play crucial roles in protecting the telomere against fusions. Herein, we discuss possible mechanisms and contributions of Ku and DNA–PKcs in telomere regulation.
Keywords DNA–PK, Shelterin, Telomerase, Telomere, hnRNP–A1
Address and Contact Information 1 Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing 400030, China
2 Department of Radiation Oncology, Sichuan Cancer Hospital, Chengdu, China.
3 Department of Radiation Oncology, University of Texas Southwestern Medical Center, 2201 Inwood Rd., Dallas, TX 75390–9187, USA
* Corresponding author: Jiangdong.Sui@hotmail.com; benjamin.chen@utsouthwestern.edu
Read full article at BMC

No. 3DOI: 10.1186/s11658-020-0200-y Volume 25 (2020) - 25:3
Title DEFICIENCY OF αII-SPECTRIN AFFECTS ENDOTHELIAL CELL–MATRIX CONTACT AND MIGRATION LEADING TO IMPAIRMENT OF ANGIOGENESIS IN VITRO
Authors Beata Machnicka1*, Aurélie Ponceau2,3,4†, Julien Picot2,3,4, Yves Colin2,3,4 and Marie-Christine Lecomte2,3,4
Abstract Background: Precise coordination of cytoskeletal components and dynamic control of cell adhesion and migration are required for crucial cell processes such as differentiation and morphogenesis. We investigated the potential involvement of αII-spectrin, a ubiquitous scaffolding element of the membrane skeleton, in the adhesion and angiogenesis mechanism.
Methods: The cell models were primary human umbilical vein endothelial cells (HUVECs) and a human dermal microvascular endothelial cell line (HMEC-1). After siRNA- and shRNA-mediated knockdown of αII-spectrin, we assessed its expression and that of its partners and adhesion proteins using western blotting. The phenotypes of the control and spectrin-depleted cells were examined using immunofluorescence and video microscopy. Capillary tube formation was assessed using the thick gel Matrigel matrix-based method and a microscope equipped with a thermostatic chamber and a Nikon Biostation System camera.
Results: Knockdown of αII-spectrin leads to: modified cell shape; actin cytoskeleton organization with the presence of peripheral actin patches; and decreased formation of stress fibers. Spectrin deficiency affects cell adhesion on laminin and fibronectin and cell motility. This included modification of the localization of adhesion molecules, such as αVβ3- and α5-integrins, and organization of adhesion structures, such as focal points. Deficiency of αII-spectrin can also affect the complex mechanism of in vitro capillary tube formation, as demonstrated in a model of angiogenesis. Live imaging revealed that impairment of capillary tube assembly was mainly associated with a significant decrease in cell projection length and stability. αII-spectrin depletion is also associated with significantly decreased expression of three proteins involved in capillary tube formation and assembly: VE-cadherin, MCAM and β3-integrin.
Conclusion: Our data confirm the role of αII-spectrin in the control of cell adhesion and spreading. Moreover, our findings further support the participation of αII-spectrin in capillary tube formation in vitro through control of adhesion molecules, such as integrins. This indicates a new function of αII-spectrin in angiogenesis.
Keywords αII-spectrin, Endothelial cells, Adhesion, Migration, Actin cytoskeleton, Capillary tube assembly
Address and Contact Information 1 University of Zielona Góra, Institute of Biological Sciences, Zielona Góra, Poland
2 Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Université de Paris, F-75015 Paris, France.
3 Institut National de la Transfusion Sanguine, F-75015 Paris, France.
4 Laboratoire d’Excellence GR-Ex, Paris, France.
* Corresponding author: b.machnicka@wnb.uz.zgora.pl
Beata Machnicka and Aurélie Ponceau contributed equally to this work.
Read full article at BMC

No. 4DOI: 10.1186/s11658-020-0201-x Volume 25 (2020) - 25:4
Title LINC00511 PROMOTES PROLIFERATION AND INVASION BY SPONGING miR-515-5p IN GASTRIC CANCER
Authors Di Wang1, Ketong Liu2 and Erdong Chen1*
Abstract Background: Long non-coding RNAs (lncRNAs) are known to be involved in tumorigenesis. The functions of LINC00511 in gastric cancer are poorly understood.
Methods: Quantitative RT-PCR was performed to investigate the levels of LINC00511 in gastric cancer tissues and cell lines. CCK-8, flow cytometry, wound-healing and Transwell assays were performed to examine cellular functions. The underlying regulatory mechanisms of LINC00511 in gastric cancer progression were determined using luciferase reporter and RIP assays.
Results: LINC00511 levels were significantly higher in gastric cancer tissues and cell lines than in normal samples. The high expression of LINC00511 in gastric cancer patient samples positively correlated with advanced clinical characters and poor prognosis. Depleting LINC00511 reduced tumor cell proliferation, migration and invasion, slowed tumor growth, and accelerated cell apoptosis. Our mechanistic study results indicated that LINC00511 promotes gastric cancer progression in a miR-515-5p-dependent manner.
Conclusion: We established that LINC00511 may contribute to the proliferation and invasion of gastric cancer cells by modulating miR-515-5p, indicating that LINC00511 may be a potential molecular target for the development of anti-cancer drugs.
Keywords Gastric cancer, LINC00511, miR-515-5p, ceRNA
Address and Contact Information 1 Department of Gastroenterology I, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, Heilongjiang, China
2 Department of Cardiology III, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161000, Heilongjiang, China.
* Corresponding author: erdongcheneast@163.com
Read full article at BMC

No. 5DOI: 10.1186/s11658-020-0202-9 Volume 25 (2020) - 25:5
Title MicroRNA-429 INHIBITS NEUROBLASTOMA CELL PROLIFERATION, MIGRATION AND INVASION VIA THE NF-κB PATHWAY
Authors Xianjun Zhou1, Hongting Lu1, Fujiang Li1, Xiwei Hao1, Lulu Han2, Qian Dong1* and Xin Chen1*
Abstract Background: MicroRNAs (miRNAs or miRs) can participate in the development and progression of neuroblastoma. Many studies have indicated that miR-429 can participate in tumor development. However, the mechanism underlying miR-429-mediated progression of neuroblastoma remains largely unclear.
Methods: Colony formation and apoptosis assays were used to determine the effect of miR-429 on cell proliferation. Its impact on cell migration was determined using the wound-healing and Transwell assays. The target gene of miR-429 was confirmed via western blotting and luciferase reporter assays. A nude mouse xenograft model with miR-429 overexpression was used to assess the effect on tumor growth.
Results: Our findings indicate that miR-429 is downregulated in neuroblastoma cell lines. We also found that it can induce apoptosis and inhibit proliferation in cells of those lines. MiR-429 can bind to the 3′-UTR of IKKβ mRNA and overexpression of IKKβ can reverse cell proliferation, blocking the effect of miR-429. Furthermore, miR-429 overexpression inhibited neuroblastoma growth in our nude mouse xenograft model.
Conclusion: We provide important insight into miR-429 as a tumor suppressor through interaction with IKKβ, which is a catalytic subunit of the IKK complex that activates NF-κB nuclear transport. Our results demonstrate that miR-429 may be a new target for the treatment of neuroblastoma.
Keywords Neuroblastoma, NF-κB signaling, miR-429, IKKβ
Address and Contact Information 1 Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266000, Shandong, China
2 Department of Operation Room, the Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao 266000, Shandong, China
* Corresponding author: 18661801885@163.com; 15506399877@163.com
Read full article at BMC

No. 6DOI:10.1186/s11658-020-00203-2 Volume 25 (2020) - 25:6
Title INHIBITION OF microRNA-15 PROTECTS H9c2 CELLS AGAINST CVB3-INDUCED MYOCARDIAL INJURY BY TARGETING NLRX1 TO REGULATE THE NLRP3 INFLAMMASOME
Authors Ru Tong1, Tiewen Jia1, Ruijie Shi2 and Futang Yan2*
Abstract Background: Viral myocarditis (VMC) is a type of cardiac inflammation that is generally caused by coxsackievirus B3 (CVB3) infection. Several MicroRNAs (miRNAs) are known to play crucial roles in VMC pathogenesis. MiR-15 is reportedly associated with myocardial injury, inflammatory responses and viral infection. Whether miR-15 affects the occurrence and development of VMC remains largely unknown. The roles of miR-15 and their underlying mechanisms in CVB3-stimulated H9c2 cells were assessed in this study.
Methods: We infected H9c2 cells with CVB3 to establish a VMC cellular model. We then determined the effects of miR-15 inhibition on three cardiomyocyte injury markers: lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB) and cardiac troponin-I (cTn-I). The impact on CVB3-induced cell apoptosis and pro-inflammatory cytokines was also investigated. The effects of miR-15 inhibition on NLRP3 inflammasome activation were also assessed. The target relationship between miR-15 and NOD-like receptor X1 (NLRX1) was determined using a luciferase reporter assay.
Results: MiR-15 expression was significantly upregulated in H9c2 cells after CVB3 infection. Inhibition of miR-15 significantly decreased the CVB3-induced levels of LDH, CK-MB and cTn-I. It also elevated cell viability, reduced CVB3-induced cell apoptosis and decreased the generation of the interleukins IL-1β, IL-6 and IL-18. Furthermore, we determined that miR-15 inhibition suppressed NLRP3 inflammasome activation by downregulating NLRP3 and caspase-1 p20 expression. We found a direct target relationship between miR-15 and NLRX1. Additionally, inhibition of NLRX1 reversed the protective effects of miR-15 inhibition against CVB3-induced myocardial cell injury by regulating the NLRP3 inflammasome.
Conclusion: Our results indicate that miR-15 inhibition alleviates CVB3-induced myocardial inflammation and cell injury. This may be partially due to NLRX1-mediated NLRP3 inflammasome inactivation.
Keywords Coxsackievirus B3, Viral myocarditis, MiR-15, NLRX1, NLRP3 inflammasome
Address and Contact Information 1 Laboratory Dept., Second Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China.
2 Laboratory Dept., Shaanxi Provincial People’s Hospital, No. 256, West Youyi Road, Xi’an 710068, Shaanxi province, China
* Corresponding author: futang_yanxa@163.com
Read full article at BMC

No. 7DOI: 10.1186/s11658-020-00204-1 Volume 25 (2020) - 25:8
Title GNAS PROMOTES INFLAMMATION-RELATED HEPATOCELLULAR CARCINOMA PROGRESSION BY PROMOTING STAT3 ACTIVATION
Authors Hongda Ding1, Xixia Zhang2, Yang Su1, Changjun Jia1 and Chaoliu Dai1*
Abstract Background: Hepatocellular carcinoma (HCC) is still the most common cause of cancer-related mortality worldwide and accumulating studies report that HCC is frequently linked to chronic inflammation. G-protein alpha-subunit (GNAS)-activating mutations have recently been reported to form a rare subgroup of inflammatory liver tumors. In this study, we investigated the roles of GNAS in inflammation-related HCC progression and its underlying mechanism.
Methods: Lipopolysaccharides (LPS) and diethylnitrosamine were employed to stimulate HCC cells to an induced inflammatory response. qRT-PCR, immunohistochemistry and immunoblotting were performed to detect the expression of GNAS in HCC tissues and cell lines. Expression levels of proinflammatory cytokines were detected by qRT-PCR and ELISA. N6-methyladenosine (m6A) methylation of GNAS mRNA was detected by RNA-binding protein immunoprecipitation (RIP). Transcription factors activation profiling plate array was performed to investigate the underlying mechanism in GNAS promoting interleukin-6 (IL-6) expression in HCC cells. HCC cell invasion was determined by transwell assay in vitro, and tumorigenesis was assessed with a subcutaneous xenograft mouse model of HCC.
Results: We found that LPS stimulation promotes GNAS expression in HCC cells through increasing m6A methylation of GNAS mRNA. The high expression level of GNAS promotes LPS-induced HCC cell growth and invasion by interacting with signal transducer and activator of transcription 3 (STAT3). Furthermore, GNAS knockdown inhibits LPS induced-IL-6 expression in HCC cells by suppressing STAT3 activation. Moreover, we found that GNAS promotes LPS-induced STAT3 activation in HCC cells through inhibiting long non-coding RNA TPTEP1 interacting with STAT3. In addition, GNAS expression promotes HCC development in mice and is related to poor survival.
Conclusions: Our findings for the first time indicate a tumor-promoting role of GNAS in inflammation-related HCC progression and provide a novel potential target for HCC therapy.
Keywords Hepatocellular carcinoma, Lipopolysaccharides, G-protein alpha-subunit, Signal transducer and activator of transcription 3, Interleukin-6
Address and Contact Information 1 Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Road, Shenyang 110004, China
2 Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Road, Shenyang 110004, China.
* Corresponding author: daiclsjh@163.com
Read full article at BMC

No. 8DOI: 10.1186/s11658-020-00205-0 Volume 25 (2020) - 25:10
Title FERROSTATIN-1 ALLEVIATES LIPOPOLYSACCHARIDE-INDUCED ACUTE LUNG INJURY VIA INHIBITING FERROPTOSIS
Authors Pengfei Liu1,2* , Yetong Feng3, Hanwei Li1,4, Xin Chen5, Guangsuo Wang6, Shiyuan Xu4, Yalan Li7,2* and Lei Zhao1,2*
Abstract Background: Ferroptosis is a newly recognized type of cell death, which is different from traditional necrosis, apoptosis or autophagic cell death. However, the position of ferroptosis in lipopolysaccharide (LPS)-induced acute lung injury (ALI) has not been explored intensively so far. In this study, we mainly analyzed the relationship between ferroptosis and LPS-induced ALI.
Methods: In this study, a human bronchial epithelial cell line, BEAS-2B, was treated with LPS and ferrostatin-1 (Fer-1, ferroptosis inhibitor). The cell viability was measured using CCK-8. Additionally, the levels of malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), and iron, as well as the protein level of SLC7A11 and GPX4, were measured in different groups. To further confirm the in vitro results, an ALI model was induced by LPS in mice, and the therapeutic action of Fer-1 and ferroptosis level in lung tissues were evaluated.
Results: The cell viability of BEAS-2B was down-regulated by LPS treatment, together with the ferroptosis markers SLC7A11 and GPX4, while the levels of MDA, 4-HNE and total iron were increased by LPS treatment in a dose-dependent manner, which could be rescued by Fer-1. The results of the in vivo experiment also indicated that Fer-1 exerted therapeutic action against LPS-induced ALI, and down-regulated the ferroptosis level in lung tissues.
Conclusions: Our study indicated that ferroptosis has an important role in the progression of LPS-induced ALI, and ferroptosis may become a novel target in the treatment of ALI patients.
Keywords Ferrostatin-1, Ferroptosis, Lipopolysaccharide, Acute lung injury
Address and Contact Information 1 Department of Anesthesiology, The 2nd Clinical Medical College (Shenzhen People’s Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen 518020, China.
2 Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China.
3 Health Science Center, School of Basic Medical Sciences, Shenzhen University, Shenzhen 518037, China.
4 Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou 510280, China.
5 Department of Laboratory Medicine, The 2nd Clinical Medicine College (Shenzhen People’s Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen 518020, China.
6 Department of Thoracic Surgery, The 2nd Clinical Medicine College (Shenzhen People’s Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen 518020, China.
7 Department of Anesthesiology, First Affiliated Hospital of Jinan University, Guangzhou 510632, China
* Corresponding author: pengfeiliu2019@sina.com; lyalan@jnu.edu.cn; zhao.lei@szhospital.com
Read full article at BMC

No. 9DOI: 10.1186/s11658-020-00207-y Volume 25 (2020) - 25:12
Title MicroRNA-125b EXERTS ANTITUMOR FUNCTIONS IN CUTANEOUS SQUAMOUS CELL CARCINOMA BY TARGETING THE STAT3 PATHWAY
Authors Ke Tian1, Wanggen Liu2, Jing Zhang3, Xiaoyi Fan3, Jingyuan Liu3, Nan Zhao3, Chunxia Yao3* and Guoying Miao1*
Abstract Background: MicroRNA-125b (miR-125b) is downregulated in human cutaneous squamous cell carcinoma (CSCC). However, its function in CSCC has yet to be extensively explored. Here, we analyze the relationship between signal transducer and activator of transcription 3 (STAT3) and miR-125b in CSCC.
Methods: Western blotting and quantitative RT-PCR were used to determine the expression of the miR-125b–STAT3 axis in human CSCC tissues and cell lines. The direct regulatory effect of miR-125b on STAT3 expression was assessed using a luciferase reporter gene assay and RNA immunoprecipitation assay. The MTT assay and flow cytometry were used to determine the role of the miR-125b–STAT3 axis in CSCC cell proliferation and apoptosis.
Results: MiR-125b expression levels were significantly lower in CSCC cell lines and tissues than in normal cell lines and tissues. STAT3 was identified as the direct target of miR-125b. Upregulation of miR-125b and downregulation of STAT3 suppressed cell proliferation and promoted cell apoptosis. Cyclin D1 and Bcl2 were identified as the downstream targets of the miR-125–STAT3 axis.
Conclusions: Our findings indicate that miR-125b acts as a tumor suppressor in CSCC by targeting the STAT3 pathway. This observation increases our understanding of the molecular mechanisms of CSCC. Therapies aimed at activating miR-125b or inhibiting STAT3 signaling should be explored as potential treatments for CSCC.
Keywords microRNA-125b, Signal transducer and activator of transcription (STAT) 3, Cutaneous squamous cell carcinoma
Address and Contact Information 1 Department of Dermatology, Affiliated Hospital of Hebei University of Engineering, Handan 056002, China.
2 Department of Histology and Embryology, Preclinical Medicine College, Xinxiang Medical University, Xinxiang 453003, China.
3 Department of Pathology, Medical School, Hebei University of Engineering, Handan 056002, China.
* Corresponding author: yaoyao20050208@163.com; guoyingmiao@163.com
Read full article at BMC

No. 10DOI: 10.1186/s11658-020-00211-2 Volume 25 (2020) - 25:15
Title METABOLIC HOST RESPONSE AND THERAPEUTIC APPROACHES TO INFLUENZA INFECTION
Authors Mohsen Keshavarz1, Farid Solaymani-Mohammadi2, Haideh Namdari3, Yaser Arjeini4, Mohammad Javad Mousavi5,6 and Farhad Rezaei4,7*
Abstract Based on available metabolomic studies, influenza infection affects a variety of cellular metabolic pathways to ensure an optimal environment for its replication and production of viral particles. Following infection, glucose uptake and aerobic glycolysis increase in infected cells continually, which results in higher glucose consumption. The pentose phosphate shunt, as another glucose-consuming pathway, is enhanced by influenza infection to help produce more nucleotides, especially ATP. Regarding lipid species, following infection, levels of triglycerides, phospholipids, and several lipid derivatives undergo perturbations, some of which are associated with inflammatory responses. Also, mitochondrial fatty acid β-oxidation decreases significantly simultaneously with an increase in biosynthesis of fatty acids and membrane lipids. Moreover, essential amino acids are demonstrated to decline in infected tissues due to the production of large amounts of viral and cellular proteins. Immune responses against influenza infection, on the other hand, could significantly affect metabolic pathways. Mainly, interferon (IFN) production following viral infection affects cell function via alteration in amino acid synthesis, membrane composition, and lipid metabolism. Understanding metabolic alterations required for influenza virus replication has revealed novel therapeutic methods based on targeted inhibition of these cellular metabolic pathways.
Keywords Influenza, Glycolysis, Fatty acid synthesis, Metabolism, Indoleamine-2,3-dioxygenase
Address and Contact Information 1 The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran.
2 Department of Biological Sciences, North Dakota State University, Fargo, North Dakota, USA.
3 Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran.
4 Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
5 Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
6 Department of Immunology and Allergy, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.
7 National Influenza Center, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
* Corresponding author: rezaie@tums.ac.ir
Read full article at BMC

No. 11DOI:10.1186/s11658-020-00209-w Volume 25 (2020) - 25:16
Title ADIPOSE-DERIVED STROMAL CELLS IN REGULATION OF HEMATOPOIESIS
Authors Jing Zhang1, Yunsheng Liu2, Wen Yin1 and Xingbin Hu1*
Abstract Over the past decade, mesenchymal stromal cells (MSCs) found in the bone marrow microenvironment have been considered to be important candidates in cellular therapy. However, the application of MSCs in clinical settings is limited by the difficulty and low efficiency associated with the separation of MSCs from the bone marrow. Therefore, distinct sources of MSCs have been extensively explored. Adipose-derived stromal cells (ASCs), a cell line similar to MSCs, have been identified as a promising source. ASCs have become increasingly popular in many fields, as they can be conveniently extracted from fat tissue. This review focuses on the properties of ASCs in hematopoietic regulation and the underlying mechanisms, as well as the current applications and future perspectives in ASC-based therapy.
Keywords Mesenchymal stromal cells, Hematopoiesis, Adipose-derived stromal cells, Therapy, Stem cells
Address and Contact Information 1 Department of Transfusion Medicine, Xijing Hospital, Xi’an 710032, China.
2 Department of Rocket Force Medicine, Third Military Medical University, Chongqing 400038, China.
* Corresponding author: hxbyqh@fmmu.edu.cn
Read full article at BMC

No. 12DOI: 10.1186/s11658-020-00208-x Volume 25 (2020) - 25:17
Title Blockage of O-linked GlcNAcylation induces AMPK-dependent autophagy in bladder cancer cells
Authors Lu Jin, Feng Yuan, Guangcheng Dai, Qiu Yao, Han Xiang, Lixia Wang, Boxin Xue, Yuxi Shan and Xiaolong Liu*
Abstract Background: High levels of the post-translational modification O-GlcNAcylation (O-GlcNAc) are found in multiple cancers, including bladder cancer. Autophagy, which can be induced by stress from post-translational modifications, plays a critical role in maintaining cellular homeostasis and regulating tumorigenesis. The impact of O-GlcNAcylation on autophagy in bladder cancer remains unclear. Here, we evaluate the change in autophagic activity in response to O-GlcNAcylation and explore the potential mechanisms.
Methods: O-GlcNAcylation levels in bladder cancer cells were altered through pharmacological or genetic manipulations: treating with 6-diazo-5-oxo-norleucine (DON) or thiamet-G (TG) or up- and downregulation of O-GlcNAc transferase (OGT) or O-GlcNAcase (OGA). Autophagy was determined using fluorescence microscopy and western blotting. Co-immunoprecipitation (Co-IP) assays were performed to evaluate whether the autophagy regulator AMP-activated protein kinase (AMPK) was O-GlcNAc modified.
Results: Cellular autophagic flux was strikingly enhanced as a result of O-GlcNAcylation suppression, whereas it decreased at high O-GlcNAcylation levels. Phosphorylation of AMPK increased after the suppression of O-GlcNAcylation. We found that O-GlcNAcylation of AMPK suppressed the activity of this regulator, thereby inhibiting ULK1 activity and autophagy.
Conclusion: We characterized a new function of O-GlcNAcylation in the suppression of autophagy via regulation of AMPK.
Keywords O-GlcNAcylation, Autophagy, ULK1, AMPK
Address and Contact Information Department of Urology, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou 215004, R.P, China
* Corresponding author: ljurol@163.com
Lu Jin and Feng Yuan contributed equally to this work.
Read full article at BMC

No. 13DOI: 10.1186/s11658-020-00212-1 Volume 25 (2020) - 25:18
Title UNFOLDED PROTEIN RESPONSE (UPR) INTEGRATED SIGNALING NETWORKS DETERMINE CELL FATE DURING HYPOXIA
Authors Sylwia Bartoszewska1* and James F. Collawn2*
Abstract During hypoxic conditions, cells undergo critical adaptive responses that include the up-regulation of hypoxia-inducible proteins (HIFs) and the induction of the unfolded protein response (UPR). While their induced signaling pathways have many distinct targets, there are some important connections as well. Despite the extensive studies on both of these signaling pathways, the exact mechanisms involved that determine survival versus apoptosis remain largely unexplained and therefore beyond therapeutic control. Here we discuss the complex relationship between the HIF and UPR signaling pathways and the importance of understanding how these pathways differ between normal and cancer cell models.
Keywords ER-stress, Angiogenesis, Hypoxia-reoxygenation injury, Ischemia, Cell fate determination, UPRmt
Address and Contact Information 1 Department of Inorganic Chemistry, Medical University of Gdansk, Gdansk, Poland
2 Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
* Corresponding author: sylwiabart@gumed.edu.pl; jcollawn@uab.edu
Read full article at BMC

No. 14DOI: 10.1186/s11658-020-00210-3 Volume 25 (2020) - 25:19
Title miR-144-3p INHIBITS CELL PROLIFERATION OF COLORECTAL CANCER CELLS BY TARGETING BCL6 VIA INHIBITION OF Wnt/β-CATENIN SIGNALING
Authors Naihui Sun1, Liang Zhang1, Chongguang Zhang1 and Yuan Yuan2*
Abstract Background: It has been shown that miR-144-3p regulates cell proliferation, apoptosis, migration and invasion in various cancers. However, the function and expression of miR-144-3p in colorectal cancer (CRC) remained obscure.
Methods: Immunohistochemical (IHC) staining was performed to investigate the protein expression of BCL6 in CRC tissues. The effect of BCL6 and miR-144-3p on CRC cells was explored through methylthiazolyl tetrazolium (MTT) assay, colony formation and cell cycle assays. Luciferase reporter assays, reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot assay were carried out to determine that BCL6 is directly regulated by miR-144-3p.
Results: Our results showed that miR-144-3p is down-regulated in CRC and correlates with the tumor progression of CRC patients. miR-144-3p inhibits cell proliferation and delays G1/S phase transition of CRC cells. Moreover, we found that BCL6 is a new target of miR-144-3p. Furthermore, BCL6 is a mediator of miR-144-3p repression of cell proliferation and cell cycle arrest in CRC cells. miR-144-3p repression of Wnt/β-catenin signaling is mediated by BCL6 in CRC cells.
Conclusions: Overall, the effect of the miR-144-3p/BCL6 axis on regulating CRC carcinogenesis was demonstrated, and miR-144-3p was identified as a potential prognostic and therapeutic target in colorectal cancer.
Keywords miRNA, miR-144-3p, BCL6, Wnt/β-catenin, CRC
Address and Contact Information 1 Department of Surgery, Ninghe Hospital, Tianjin 301500, China.
2 Department of Retired Office, North China University of Science and Technology, No. 46, Xinhuaxi road, Tangshan 063000, Hebei, China.
* Corresponding author: zcyzcbyy@163.com
Read full article at BMC

No. 15DOI: 10.1186/s11658-020-00213-0 Volume 25 (2020) - 25:20
Title DOWNREGULATION OF miR-483-5p DECREASES HYPOXIA-INDUCED INJURY IN HUMAN CARDIOMYOCYTES BY TARGETING MAPK3
Authors Yan Hao, Haitao Yuan* and Houzhi Yu*
Abstract Background: MiR-483-5p was recently identified as a risk factor in the early stages of acute myocardial infarction (AMI) patients. Here, we further investigated how miR-483-5p affects cardiomyocyte apoptosis and oxidative stress under hypoxic conditions.
Methods: Plasma samples were collected from AMI patients and healthy volunteers. The expression of miR-483-5p was determined using quantitative real-time PCR. An in vitro hypoxic model was constructed to mimic AMI in AC16 cells. Cell viability, apoptosis and oxidative stress biomarker levels (MDA, SOD and CAT) were respectively determined using CCK-8, flow cytometry and commercial assay kits.
Results: The expression levels of miR-483-5p were significantly higher in AMI patients than in control subjects. Circulating levels of miR-483-5p positively correlated with creatine kinase MB isoform (CK-MB) and cardiac troponin I (cTnI) levels. The in vitro experiments showed that the expression levels of miR-483-5p were also upregulated in hypoxia-induced AC16 cell injury. MiR-483-5p overexpression significantly increased hypoxia-induced cardiomyocyte apoptosis and oxidative stress, while knockdown attenuated these effects. Mechanistically, miR-483-5p directly targets MAPK3 in AC16 cells. Furthermore, the protective effects of miR-483-5p knockdown against hypoxia-induced cardiomyocyte injury are partially dependent on MAPK3.
Conclusions: MiR-483-5p, which targets MAPK3, might be a potential therapeutic target for the diagnosis and prevention of hypoxia-induced myocardial injury.
Keywords Acute myocardial infarction, miR-483-5p, MAPK3, Apoptosis, Oxidative stress
Address and Contact Information Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan 250021, Shandong, China
* Corresponding author: yuan_haitao0204@126.com; yeuosjdliyuaj23@2980.com
Read full article at BMC

No. 16DOI: 10.1186/s11658-020-00215-y Volume 25 (2020) - 25:21
Title LncRNA CASC2 INHIBITS HYPOXIA-INDUCED PULMONARY ARTERY SMOOTH MUSCLE CELL PROLIFERATION AND MIGRATION BY REGULATING THE miR-222/ING5 AXIS
Authors Yan Han, Yuhao Liu, Chaokuan Yang, Chuanyu Gao, Xiaoyan Guo and Jiangtao Cheng*
Abstract Background: Pulmonary arterial hypertension (PAH) is often characterized by cell proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs). LncRNA cancer susceptibility candidate 2 (CASC2) has been revealed to be involved in PASMC injury in hypoxia-induced pulmonary hypertension. However, the exact molecular mechanisms whereby CASC2 regulates PASMC proliferation and migration are still incompletely understood.
Methods: The expression levels of CASC2, miR-222 and inhibitor of growth 5 (ING5) were measured using quantitative real-time polymerase chain reaction (qRT-PCR) or western blot, respectively. Cell proliferation was analyzed by Cell Counting Kit-8 (CCK-8) assay. Wound healing assay was used to analyze cell migration ability. The relationship between miR-222 and CASC2 or ING5 was confirmed using bioinformatics analysis, luciferase reporter assay and RNA immunoprecipitation assay.
Results: CASC2 was down-regulated in hypoxia-induced PASMCs in a dose- and time-dependent manner. Functional experiments showed that CASC2 overexpression could reverse hypoxia-induced proliferation and migration of PASMCs. Bioinformatics analysis indicated that CASC2 acted as a competing endogenous RNA of miR-222, thereby regulating the expression of ING5, the downstream target of miR-222, in PASMCs. In addition, rescue assay suggested that the inhibition mediated by CASC2 of hypoxia-induced PASMC proliferation and migration could be attenuated by miR-222 inhibition or ING5 overexpression.
Conclusion: CASC2 attenuated hypoxia-induced PASMC proliferation and migration by regulating the miR-222/ING5 axis to prevent vascular remodeling and the development of PAH, providing a novel insight and therapeutic strategy for hypoxia-induced PAH.
Keywords PAH, CASC2, miR-222, ING5, Proliferation, Migration
Address and Contact Information Department of Cardiology, Henan Province People’s Hospital, Huazhongfuwai Hospital, No. 7, Weiwu Road, Jinshui area, Zhengzhou City, Henan, P.R. China
* Corresponding author: zhijing65820688@126.com,
Yan Han and Yuhao Liu contributed equally to this work.
Read full article at BMC

No. 17DOI: 10.1186/s11658-020-00216-x Volume 25 (2020) - 25:22
Title MiR-206 MAY SUPPRESS NON-SMALL LUNG CANCER METASTASIS BY TARGETING CORO1C
Authors Ming Liao and Lijun Peng*
Abstract Object: Non-small lung cancer (NSCLC), with a poor 5-year survival rate (16%), is the major type of lung cancer. Metastasis has been identified as the main factor that leads to NSCLC therapy failure. MiR-206 is a metastasis suppressor in many cancers, including colorectal cancer, renal cell carcinoma and breast cancer. However, the role of miR-206 in NSCLC metastasis and the underlying mechanism are still obscure.
Methods: Quantitative reverse-transcription PCR (q-RT-PCR) assay was used to detect miR-206 mRNA of NSCLC tissues and lung cancer lines. The MTT assay, scratch wound healing assay, transwell migration assay and transwell invasion assay were conducted to illuminate the effect of miR-206 on A549 cells’ proliferation, migration and invasion. Gaussia luciferase reporter assay, q-RT-PCR and western blotting assay were used to explore the underlying mechanism. Also, the A549 xenograft model was conducted to evaluate the anti-tumor effect of miR-206 in vivo.
Results: The results showed that miR-206 expression was decreased in NSCLC tissues and lung cancer cells. Further research demonstrated that miR-206 inhibited the proliferation, migration and invasion of A549 cells via negatively regulating Coronin-1C (CORO1C), and CORO1C deletion significantly rescues the miR-206 mediated inhibitory effect on A549 cells. Moreover, miR-206 exhibited a perfect anti-tumor effect in the A549 xenograft model.
Conclusion: Our study reveals that miR-206 functions as a tumor metastasis suppressor and sheds new light on the clinical significance of miR-206 in NSCLC therapy.
Keywords Non-small-cell lung cancer (NSCLC), microR-206 (miR-206), Proliferation, Migration and invasion, Coronin-1C (CORO1C)
Address and Contact Information Thoracic Surgery Department, General Hospital of Southern Theater Command, PLA, No. 111, Liuhua Road, Yuexiu District, Guangzhou 510010, China
* Corresponding author: DOCTORPL@163.com
Read full article at BMC

No. 18DOI: 10.1186/s11658-020-00214-z Volume 25 (2020) - 25:23
Title THE ROLE OF PHOSPHATIDYLSERINE RECOGNITION RECEPTORS IN MULTIPLE BIOLOGICAL FUNCTIONS
Authors Mehri Bemani Naeini1, Vanessa Bianconi2, Matteo Pirro2 and Amirhossein Sahebkar3,4,5*
Abstract Apoptotic cells are rapidly engulfed and degraded by phagocytes through efferocytosis. Efferocytosis is a highly regulated process. It is triggered upon the activation of caspase-dependent apoptosis, which in turn promotes the expression of “eat me” signals on the surface of dying cells and the release of soluble “find me” signals for the recruitment of phagocytes. To date, many “eat me” signals have been recognized, including phosphatidylserine (PS), intercellular adhesion molecule-3, carbohydrates (e.g., amino sugars, mannose) and calreticulin. Among them, PS is the most studied one. PS recognition receptors are different functionally active receptors expressed by phagocytes. Various PS recognition receptors with different structure, cell type expression, and ability to bind to PS have been recognized. Although PS recognition receptors do not fall into a single classification or family of proteins due to their structural differences, they all share the common ability to activate downstream signaling pathways leading to the production of anti-inflammatory mediators. In this review, available evidence regarding molecular mechanisms underlying PS recognition receptor-regulated clearance of apoptotic cells is discussed. In addition, some efferocytosis-independent biological functions of PS recognition receptors are reviewed.
Keywords Apoptosis, Efferocytosis, Macrophage, Phosphatidylserine, Phosphatidylethanolamine, Receptor
Address and Contact Information 1 Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
2 Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy.
3 Halal Research Center of IRI, FDA, Tehran, Iran.
4 Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
5 Department of Medical Biotechnology, School of Medicine, Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, P.O. Box: 91779-48564, Mashhad, Iran.
* Corresponding author: sahebkara@mums.ac.ir; amir_saheb2000@yahoo.com
Read full article at BMC

No. 19DOI: 10.1186/s11658-020-00217-w Volume 25 (2020) - 25:24
Title TCF 4 TUMOR SUPPRESSOR: A MOLECULAR TARGET IN THE PROGNOSIS OF SPORADIC COLORECTAL CANCER IN HUMANS
Authors Mumtaz Anwar1,2,3, Pooja Malhotra2,4, Rakesh Kochhar2, Alka Bhatia1, Akhtar Mahmood5, Rajinder Singh6 and Safrun Mahmood1*
Abstract Background: A huge array of function is played by the Wnt/β-catenin signaling pathway in development by balancing gene expression through the modulation of cell-specific DNA binding downstream effectors such as T-cell factor/lymphoid enhancer factor (TCF/LEF). The β-catenin/TCF-4 complex is a central regulatory switch for differentiation and proliferation of intestinal cells (both normal and malignant). Thus, in the present study we evaluated each of 60 cases of sporadic adenocarcinoma, alongside adjoining and normal mucosa specimens of colorectum in humans, for mutation and expression analysis of the gene coding for TCF-4 protein.
Methods: DNA sequencing following PCR amplification and SSCP analysis (single strand conformation polymorphism) was employed to detect TCF-4 gene mutations in the case of exon 1. Quantitative real-time (qRT) PCR, immunohistochemistry (IHC), confocal microscopy and western blot analysis were used to detect TCF-4 gene/protein expression.
Results: Sequencing analysis confirmed 5/60 patients with a point mutation in exon 1 of the TCF-4 gene in tumor samples. mRNA expression using qRT-PCR showed approximately 83% decreased TCF-4 mRNA expression in tumor tissue and adjoining mucosa compared to normal mucosa. Similarly, a significant decrease in protein expression using IHC showed decreased TCF-4 protein expression in tumor tissue and adjoining mucosa compared to normal mucosa, which also corresponds to some important clinicopathological factors, including disease metastasis and tumor grade. Mutational alterations and downregulation of TCF-4 mRNA and hence decreased expression of TCF-4 protein in tumors suggest its involvement in the pathogenesis of CRC.
Conclusions: A remarkable decrease in TCF-4 mRNA and protein expression was detected in tumorous and adjoining tissues compared to normal mucosa. Hence the alterations in genomic architecture along with downregulation of TCF-4 mRNA and decreased expression of TCF-4 protein in tumors, which is in accordance with clinical features, suggest its involvement in the pathogenesis of CRC. Thus, deregulation and collaboration of TCF-4 with CRC could be a concrete and distinctive feature in the prognosis of the disease at an early stage of development.
Keywords TCF-4, Genetic alterations, Quantitative real-time PCR, IHC, Confocal microscopy, Western blotting, Clinicopathological factors
Address and Contact Information 1 Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh 160012, India
2 Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India.
3 Department of Pharmacology, University of Illinois at Chicago, Chicago 60612, USA.
4 Department of Medicine, University of Illinois at Chicago, Chicago 60612, USA.
5 Department of Biochemistry, Panjab University, Chandigarh 160014, India.
6 Department of Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India.
* Corresponding author: mahmoodpgi@gmail.com
Read full article at BMC

No. 20DOI: 10.1186/s11658-020-00218-9 Volume 25 (2020) - 25:25
Title QUANTITATIVE PROTEOMICS ANALYSIS OF DIFFERENTIALLY EXPRESSED PROTEINS INDUCED BY ASTRAGALOSIDE IV IN CERVICAL CANCER CELL INVASION
Authors Chenglai Xia1*, Zhihong He1 and Yantao Cai2
Abstract Background: Cervical cancer remains the second leading cause of mortality in women in developing countries. While surgery, chemotherapy, radiotherapy, and vaccine therapy are being applied for its treatment, individually or in combination, the survival rate in advanced cervical cancer patients is still very low. Traditional Chinese medicine has been found to be effective in the treatment of cervical cancer. Astragaloside IV (AS-IV), a compound belonging to Astragalus polysaccharides, shows anticancer activity through several cell signaling pathways. However, the detailed molecular mechanism governing the anticancer activity of AS-IV remains unknown.
Material and methods: In our study, we performed tumor xenograft analysis, transwell cell migration and invasion assay, Western blot analysis, and iTRAQ combination by parallel reaction monitoring (PRM) analysis to study the molecular mechanism of AS-IV in the suppression of cervical cancer cell invasion.
Results: Our results showed that AS-IV suppressed cervical cancer cell invasion and induced autophagy in them, with the tumor growth curve increasing slowly. We also identified 32 proteins that were differentially expressed in the SiHa cells when treated with AS-IV, with 16 of them involved in the upregulation and 16 in the downregulation of these cells. These differentially expressed proteins, which were predominantly actin–myosin complexes, controlled cell proliferation and cell development by steroid binding and altering the composition of the cell cytoskeleton. DCP1A and TMSB4X, the two proteins regulating autophagy, increased in cervical cancer cells when treated with AS-IV.
Conclusions: We conclude that AS-IV could inhibit cervical cancer invasion by inducing autophagy in cervical cancer cells. Since iTRAQ combination by PRM has been observed to be useful in identifying macromolecular target compounds, it may be considered as a novel strategy in the screening of anticancer compounds used in the treatment of cervical cancer.
Keywords Astragaloside IV, Quantitative proteomics, iTRAQ, Parallel reaction monitoring, Cervical cancer
Address and Contact Information 17 Foshan Maternal and Child Health Research Institute, South Medical University Affiliated Maternal & Child Health Hospital of Foshan, 11 Renmin Xi Street, Foshan 528000, China
2 Department of Dermatology and Pheumatology, South Medical University Affiliated Maternal & Child Health Hospital of Foshan, 11 Renmin Xi Street, Foshan 528000, China.
* Corresponding author: xiachenglai@126.com
Read full article at BMC

No. 21DOI: 10.1186/s11658-020-00206-z Volume 25 (2020) - 25:26
Title THE ROLE OF miR-711 IN CARDIAC CELLS IN RESPONSE TO OXIDATIVE STRESS AND ITS BIOGENESIS: A STUDY ON H9C2 CELLS
Authors Duo Zhao1,2,3†, Hao Zheng2†, Adam Greasley2†, Fengjun Ling2, Qinfeng Zhou2,4, Bowen Wang2, Tiffany Ni2, Ishita Topiwala2, Cuilin Zhu1,2, Tina Mele5, Kexiang Liu1* and Xiufen Zheng2,5,6,7,8*
Abstract Background: Oxidative stress results in cell apoptosis/death and plays a detrimental role in disease development and progression. Stressors alter the miRNA expression profile and miRNAs play a role in the cell response to stress. We previously showed that miR-711 is significantly over-expressed in extended cold ischemia reperfusion injured hearts in heart transplant. In this study, we aimed to investigate the role of miR-711 in cardiac cell damage in response to oxidative stress and how miR-711 is regulated.
Methods: Rat cardiac cell line H9c2 cells were cultured and exposed to oxidative conditions (Antimycin A (AA), H2O2, CoCl2, or cold hypoxia/reoxygenation (H/R)) in vitro. H9c2 cells were transfected with miR-711 mimics, miR-711 inhibitors, or small interference RNA, using transfection reagents. The expression of miR-711 was measured by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). Cell apoptosis/death was detected by flow cytometry and an IncuCyte system. Mitochondrial damage was detected by measuring the mitochondria membrane potential by flow cytometry. Gene expression was detected by qRT-PCR at the mRNA level and Western blotting and immunocytochemistry staining at the protein level.
Results: We found that miR-711 was significantly up-regulated in cells treated with H2O2, AA, CoCl2, and cold H/R. Over-expression of miR-711 increased cell apoptosis/death induced by AA and H/R whereas cell death was reduced by miR-711 inhibitors. MiR-711 induced cell death through negative regulation of angiopoietin 1 (Ang-1), fibroblast growth factor 14 (FGF14) and calcium voltage-gated channel subunit alpha1C (Cacna1c) genes. Both knockdown of hypoxia inducible factor 1α (HIF-1α) and inactivation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NFКB) pathway inhibited over-expression of miR-711.
Conclusion: Oxidative stress increases the expression of miR-711. Over-expression of miR-711 induces cell apoptosis/death. HIF-1α and NFКB regulate miR-711 in H9c2 cells during oxidative stress. miR-711 is a new target for preventing oxidative stress.
Keywords miR-711, Oxidative stress, HIF-1α, NFКB, Ang-1, FGF14, Cacna1c
Address and Contact Information 1 Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun 130041, China
2 Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
3 Department of Cardiovascular Surgery, The First People’s Hospital of Foshan, Foshan, Guangdong, China.
4 Department of Laboratory Medicine, Zhangjiagang TCM Hospital Affiliated to Nanking University of Chinese Medicine, Zhangjiagang, Jiangsu, China.
5 Department of Surgery, Western University, Ontario, London, Canada.
6 London Health Sciences Centre, London, Ontario, Canada.
7 Department of Oncology, Western University, Ontario, London, Canada.
8 Lawson Health Research Institute, Ontario, London, Canada.
* Corresponding author: kxliu1964@hotmail.com; xzheng26@uwo.ca
Duo Zhao, Hao Zheng and Adam Greasley contributed equally to this work.
Read full article at BMC

No. 22DOI: 10.1186/s11658-020-00220-1 Volume 25 (2020) - 25:27
Title MicroRNA-186-5p SERVES AS A DIAGNOSTIC BIOMARKER IN ATHEROSCLEROSIS AND REGULATES VASCULAR SMOOTH MUSCLE CELL PROLIFERATION AND MIGRATION
Authors Bin Sun1, Qingtao Cao2, Meng Meng2 and Xiaolong Wang3*
Abstract Objective: MicroRNA dysregulation occurs in many human diseases, including atherosclerosis. Here, we examined the serum expression and clinical significance of miR-186-5p in patients with atherosclerosis, and explored its influence on vascular smooth muscle cell (VSMC) proliferation and migration.
Methods: Blood samples were collected from 104 patients with asymptomatic atherosclerosis and 80 healthy controls. Quantitative real-time PCR was applied to measure the miR-186-5p level. An ROC curve was established to assess the discriminatory ability of the serum miR-186-5p level for identifying atherosclerosis from controls. CCK-8 and Transwell assays were used to evaluate the impact of miR-186-5p on cell behaviors.
Results: Serum expression of miR-186-5p was significantly higher in atherosclerosis patients than in the control group. The serum miR-186-5p level showed a positive correlation with CIMT and could be used to distinguish atherosclerosis patients from healthy controls, with an area under the curve (AUC) score of 0.891. In VSMCs, overexpression of miR-186-5p significantly promoted cell proliferation and migration, while the opposite results were observed when miR-186-5p was downregulated.
Conclusion: Overexpression of miR-186-5p has a certain diagnostic significance for atherosclerosis. Upregulation of miR-186-5p stimulates VSMC proliferation and migration. Therefore, it is a possible target for atherosclerosis interventions.
Keywords MiR-186-5p, Atherosclerosis, Vascular smooth muscle cell, Proliferation, Migration
Address and Contact Information 1 Department of Emergency Medicine, Yidu Central Hospital of Weifang, Weifang 262500, Shandong, China.
2 Department of Cardiovascular Medicine, Yidu Central Hospital of Weifang, Weifang 262500, Shandong, China.
3 Department of Emergency Cardiovascular Medicine, Weifang People’s Hospital, No. 151 Guangwen Street, Weifang 261000, Shandong, China.
* Corresponding author: longliangc8276@163.com
Read full article at BMC

No. 23DOI: 10.1186/s11658-020-00219-8 Volume 25 (2020) - 25:28
Title TARGETING THE 4-1BB COSTIMULATORY MOLECULE THROUGH SINGLE CHAIN ANTIBODIES PROMOTES THE HUMAN T-CELL RESPONSE
Authors Salman Bagheri1,2,3, Elmira Safaie Qamsari1,2,3, Mehdi Yousefi2, Farhad Riazi-Rad1 and Zahra Sharifzadeh1*
Abstract Background: Adoptive T-cell therapy (ACT) using autologous tumor-reactive T lymphocytes has considerable potential for cancer immunotherapy. In ACT, T cells are isolated from cancer patients and then stimulated and expanded in vitro by cytokines and costimulatory molecules. 4-1BB is an important costimulatory protein belonging to the TNF receptor superfamily. It is involved in T-cell survival, proliferation and activation. Agonistic anti-4-1BB monoclonal antibodies have been introduced as appropriate tools for ACT.
Methods: Here, various single-chain fragment variable (scFv) antibodies were used to activate T cells isolated from peripheral blood via immune magnetic isolation. The T cells were stimulated with IL-2 and anti-CD-3 mAb and then treated with agonistic anti-4-1BB scFvs. The results showed the remarkable effects of anti-41BB scFvs on the functional properties of T cells, including their activation, proliferation and cytokine production. The flow cytometry analysis revealed a considerable increase in the expression of the T-cell activation marker CD69. Moreover, T-cell proliferation was evidenced in treated cells by CFSE labeling compared to the control groups.
Result: Anti-4-1BB scFvs significantly increased IFN-γ and IL-2 mRNA and protein expression in T cells, but exhibited no stimulatory effect on IL-4 expression. These findings show that anti-4-1BB scFvs could evoke a Type I immune response.
Conclusions: Our results demonstrate that targeting the 4-1BB molecule using agonistic scFvs could be an effective strategy for T-cell stimulation as part of an ACT approach to cancer treatment.
Keywords 4-1BB, Single-chain fragment antibody, T-cell therapy, Immunomodulation, T cell responses
Address and Contact Information 1 Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.
2 Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
3 Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
* Corresponding author: zsharifzadeh@gmail.com
Read full article at BMC

No. 24DOI: 10.1186/s11658-020-00221-0 Volume 25 (2020) - 25:29
Title LncRNA DRAIC INHIBITS PROLIFERATION AND METASTASIS OF GASTRIC CANCER CELLS THROUGH INTERFERING WITH NFRKB DEUBIQUITINATION MEDIATED BY UCHL5
Authors Zheng Zhang, Xiaoxuan Hu, Jia Kuang, Jinmao Liao and Qi Yuan*
Abstract Background: Long non-coding RNA (lncRNA) as a widespread and pivotal epigenetic molecule participates in the occurrence and progression of malignant tumors. DRAIC, a kind of lncRNA whose coding gene location is on 15q23 chromatin, has been found to be weakly expressed in a variety of malignant tumors and acts as a suppressor, but its characteristics and role in gastric cancer (GC) remain to be elucidated.
Methods: Sixty-seven primary GC tissues and paired paracancerous normal tissues were collected. Bioinformatics is used to predict the interaction molecules of DRAIC. DRAIC and NFRKB were overexpressed or interfered exogenously in GC cells by lentivirus or transient transfection. Quantitative real-time PCR (qPCR) and western blotting were used to evaluate the expression of DRAIC, UCHL5 and NFRKB. The combinations of DRAIC and NFRKB or UCHL5 and NFRKB were verified by RNA-IP and Co-IP assays. Ubiquitination-IP and the treatment of MG132 and CHX were used to detect the ubiquitylation level of NFRKB. The CCK-8 and transwell invasion and migration assays measured the proliferation, migration and invasion of GC cells.
Results: DRAIC is down-regulated in GC tissues and cell lines while its potential interacting molecules UCHL5 and NFRKB are up-regulated, and DRAIC is positively correlated with NFRKB protein instead of mRNA. Lower DRAIC and higher UCHL5 and NFRKB indicated advanced progression of GC patients. DRAIC could increase NFRKB protein significantly instead of NFRKB mRNA and UCHL5, and bind to UCHL5. DRAIC combined with UCHL5 and attenuated binding of UCHL5 and NFRKB, meanwhile promoting the degradation of NFRKB via ubiquitination, and then inhibited the proliferation and metastasis of GC cells, which can be rescued by oeNFRKB.
Conclusion: DRAIC suppresses GC proliferation and metastasis via interfering with the combination of UCHL5 and NFRKB and mediating ubiquitination degradation.
Keywords Gastric cancer, Long non-coding RNA DRAIC, Deubiquitination, UCHL5, NFRKB
Address and Contact Information Department of Hepatopathy, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410000, Hunan Province, China
* Corresponding author: yuanqi1416@126.com
Read full article at BMC

No. 25DOI: 10.1186/s11658-020-00222-z Volume 25 (2020) - 25:30
Title DOWNREGULATION OF miR-637 PROMOTES VASCULAR SMOOTH MUSCLE CELL PROLIFERATION AND MIGRATION VIA REGULATION OF INSULIN-LIKE GROWTH FACTOR-2
Authors Ning Yang, Bo Dong, Yanqiu Song, Yang Li, Lu Kou, Jingyu Yang and Qin Qin*
Abstract Background: Dysregulation of the proliferation and migration of vascular smooth muscle cells (VSMCs) is a crucial cause of atherosclerosis. MiR-637 exerts an antiproliferative effect on multiple human cells. Its impact on atherosclerosis remains largely unexplored.
Methods: Real-time PCR was used to determine miR-637 expression in samples from atherosclerosis patients and animal models. Its expression in VSMC dysfunction models (induced by ox-LDL) was also measured. The proliferation and migration of VSMCs were respectively tested using CCK-8 and Transwell assays, and apoptosis was measured using flow cytometry. The Targetscan database was used to predict the target genes of miR-637. Interaction between miR-637 and the potential target gene was validated via real-time PCR, western blotting and a luciferase reporter assay.
Results: MiR-637 expression was significantly lower in atherosclerosis patient and animal model samples. It also decreased in a dose- and time-dependent manner in animal models with ox-LDL-induced atherosclerosis. Transfection with miR-637 mimics suppressed the proliferation and migration of VSMCs while promoting apoptosis, while transfection with miR-637 inhibitors had the opposite effects. We also validated that insulin-like growth factor-2 (IGF-2), a crucial factor in the pathogenesis of atherosclerosis, serves as a target gene for miR-637.
Conclusion: MiR-637 targeting IGF-2 contributes to atherosclerosis inhibition and could be a potential target for this disease.
Keywords miR-637, IGF-2, VSMC, Atherosclerosis
Address and Contact Information Department of Cardiology, Tianjin Chest hospital, Taierzhuang South Road No.261, Jinnan District, Tianjin 300222, China
* Corresponding author: qinqintch@163.com
Read full article at BMC

No. 26DOI: 10.1186/s11658-020-00225-w Volume 25 (2020) - 25:31
Title ROLE OF ANGIOTENSIN-CONVERTING ENZYME IN MYELOID CELL IMMUNE RESPONSES
Authors Duo-Yao Cao1, Suguru Saito1, Luciana C. Veiras1, Derick Okwan-Duodu1,2, Ellen A. Bernstein1, Jorge F. Giani1,2, Kenneth E. Bernstein1,2 and Zakir Khan1,2*
Abstract Angiotensin-converting enzyme (ACE), a dicarboxypeptidase, plays a major role in the regulation of blood pressure by cleaving angiotensin I into angiotensin II (Ang II), a potent vasoconstrictor. Because of its wide substrate specificity and tissue distribution, ACE affects many diverse biological processes. In inflammatory diseases, including granuloma, atherosclerosis, chronic kidney disease and bacterial infection, ACE expression gets upregulated in immune cells, especially in myeloid cells. With increasing evidences connecting ACE functions to the pathogenesis of these acquired diseases, it is suggested that ACE plays a vital role in immune functions. Recent studies with mouse models of bacterial infection and tumor suggest that ACE plays an important role in the immune responses of myeloid cells. Inhibition of ACE suppresses neutrophil immune response to bacterial infection. In contrast, ACE overexpression in myeloid cells strongly induced bacterial and tumor resistance in mice. A detailed biochemical understanding of how ACE activates myeloid cells and which ACE peptide(s) (substrate or product) mediate these effects could lead to the development of novel therapies for boosting immunity against a variety of stimuli, including bacterial infection and tumor.
Keywords Angiotensin-converting enzyme, Myeloid cells, Immune response, Neutrophils, Macrophages, Dendritic cells, Hematopoiesis, Methicillin-resistant Staphylococcus aureus (MRSA), Melanoma, MHC class I antigen presentation
Address and Contact Information 1 Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
2 Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Davis Res. Bldg., Rm. 2014, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
* Corresponding author: zakir.khan@csmc.edu
Read full article at BMC

No. 27DOI: 10.1186/s11658-020-00223-y Volume 25 (2020) - 25:32
Title ON THE PHYSIOLOGICAL AND CELLULAR HOMEOSTASIS OF ASCORBATE
Authors Magdalena Przybyło1,2* and Marek Langner1,2
Abstract Recent interest in the role of ascorbate in crucial metabolic processes is driven by the growing number of medical reports that show beneficial effects of ascorbate supplementation for maintaining general well-being and recovery from a variety of medical conditions. The effect of ascorbate on the local body environment highly depends on its local concentration; at low concentrations it can cause the reduction of reactive oxygen and facilitate activities of enzymes, while at high concentrations it generates free radicals by reducing ferric ions. Ascorbate serving as an electron donor assists the iron-containing proteins and the iron transfer between various aqueous compartments. These functions require effective and adjustable mechanisms responsible for ascorbate biodistribution. In the paper we propose a new biophysical model of ascorbate redistribution between various aqueous body compartments. It combines recent experimental evidence regarding the ability of ascorbate to cross the lipid bilayer by unassisted diffusion, with active transport by well-characterized sodium vitamin C transporter (SVCT) membrane proteins. In the model, the intracellular concentration of ascorbate is maintained by the balance of two opposing fluxes: fast active and slow passive transport. The model provides a mechanistic understanding of ascorbate flux across the epidermal barrier in the gut as well as the role of astrocytes in ascorbate recycling in the brain. In addition, ascorbate passive diffusion across biological membranes, which depends on membrane electric potentials and pH gradients, provides the rationale for the correlation between ascorbate distribution and the transfer of iron ions inside a cell. The proposed approach provides, for the first time, a mechanistic account of processes leading to ascorbate physiological and cellular distribution, which helps to explain numerous experimental and clinical observations.
Keywords Vitamin C, Biodistribution, Membrane transport, Homeostasis, Membrane potentials
Address and Contact Information 1 Faculty of Biomedical Engineering, Wrocław University of Sciences and Technology, 50-370 Wrocław, Poland
2 Lipid Systems Ltd, Krzemieniecka 48C, 54-613 Wrocław, Poland
* Corresponding author: magdalena.przybylo@pwr.edu.pl
Read full article at BMC

No. 28DOI: 10.1186/s11658-020-00226-9 Volume 25 (2020) - 25:33
Title ASIATICOSIDE INHIBITS TGF-β1-INDUCED MESOTHELIAL-MESENCHYMAL TRANSITION AND OXIDATIVE STRESS VIA THE Nrf2/HO-1 SIGNALING PATHWAY IN THE HUMAN PERITONEAL MESOTHELIAL CELL LINE HMrSV5
Authors Junyi Zhao†, Jun Shi†, Yun Shan, Manshu Yu, Xiaolin Zhu, Yilin Zhu, Li Liu and Meixiao Sheng*
Abstract Background: Peritoneal fibrosis (PF) is a frequent complication caused by peritoneal dialysis (PD). Peritoneal mesothelial cells (PMCs), the first barrier of the peritoneum, play an important role in maintaining structure and function in the peritoneum during PD. Mesothelial-mesenchymal transition (MMT) and oxidative stress of PMCs are two key processes of PF.
Purpose: To elucidate the efficacy and possible mechanism of asiaticoside inhibition of MMT and ROS generation in TGF-β1-induced PF in human peritoneal mesothelial cells (HPMCs).
Methods: MMT and ROS generation of HPMCs were induced by TGF-β1. To explain the anti-MMT and antioxidant role of asiaticoside, varied doses of asiaticoside, oxygen radical scavenger (NAC), TGF-β receptor kinase inhibitor (LY2109761) and Nrf2 inhibitor (ML385) were used separately. Immunoblots were used to detect the expression of signaling associated proteins. DCFH-DA was used to detect the generation of ROS. Transwell migration assay and wound healing assay were used to verify the capacity of asiaticoside to inhibit MMT. Immunofluorescence assay was performed to observe the subcellular translocation of Nrf2 and expression of HO-1.
Results: Asiaticoside inhibited TGF-β1-induced MMT and suppressed Smad signaling in a dose-dependent manner. Migration and invasion activities of HPMCs were decreased by asiaticoside. Asiaticoside decreased TGF-β1-induced ROS, especially in a high dose (150 μM) for 6 h. Furthermore, ML385 partly abolished the inhibitory effect of asiaticoside on MMT, ROS and p-Smad2/3.
Conclusions: Asiaticoside inhibited the TGF-β1-induced MMT and ROS via Nrf2 activation, thus protecting the peritoneal membrane and preventing PF.
Keywords Human peritoneal mesothelial cells (HPMCs), Mesothelial-mesenchymal transition (MMT), Reactive oxygen species (ROS), TGF-β/Smad, Nrf-2/HO-1
Address and Contact Information Renal Division, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing 210029, Jiangsu Province, China
* Corresponding author: jsszyysmx@163.com
† Junyi Zhao and Jun Shi contributed equally to this work.
Read full article at BMC

No. 29DOI: 10.1186/s11658-020-00227-8 Volume 25 (2020) - 25:34
Title LncRNA NNT-AS1 PROMOTES LUNG SQUAMOUS CELL CARCINOMA PROGRESSION BY REGULATING THE miR-22/FOXM1 AXIS
Authors Jing Ma†, Guanbin Qi† and Lei Li*
Abstract Background: Recent studies have revealed that dysregulated expression of long non-coding RNA nicotinamide nucleotide transhydrogenase antisense RNA 1 (lncRNA NNT-AS1) is associated with cell tumorigenicity in non-small cell lung cancer. However, the exact molecular mechanisms of NNT-AS1 in lung squamous cell carcinoma (LUSC) remain largely unknown.
Methods: The expression of NNT-AS1, microRNA (miR)-22 and Forkhead box protein M1 (FOXM1) was measured using quantitative real-time polymerase chain reaction or western blot, respectively. The interaction between miR-22 and NNT-AS1 or FOXM1 was confirmed using a dual-luciferase reporter assay and RNA immunoprecipitation assay. Cell migration and invasion abilities were measured by Transwell assay. Flow cytometry was used to detect apoptotic cells.
Results: NNT-AS1 and FOXM1 were up-regulated but miR-22 was down-regulated in LUSC tissues and cell lines. NNT-AS1 was a sponge of miR-22, and NNT-AS1 deletion suppressed the migration and invasion but induced apoptosis in LUSC cells. FOXM1 was a target of miR-22, and overexpression of miR-22 inhibited cell carcinogenesis in LUSC by targeting FOXM1. Additionally, NNT-AS1 could directly regulate FOXM1 expression by binding to miR-22 in LUSC cells.
Conclusion: LncRNA NNT-AS1 contributes to cell carcinogenesis in LUSC by regulating the miR-22/FOXM1 axis, providing a novel insight into the pathogenesis of LUSC and a new potential therapeutic target for LUSC treatment.
Keywords NNT-AS1, miR-22, FOXM1, LUSC, Progression
Address and Contact Information Department of Respiratory and Critical Care Medicine, Huaihe Hospital of Henan University, NO.115 Ximen Street, Kaifeng City, Henan Province, Kaifeng 475000, Henan, China
* Corresponding author: qiyi3775669@yeah.net
† Jing Ma and Guanbin Qi contributed equally to this work.
Read full article at BMC

No. 30DOI: 10.1186/s11658-020-00228-7 Volume 25 (2020) - 25:35
Title THE USEFULNESS OF LACTATE DEHYDROGENASE MEASUREMENTS IN CURRENT ONCOLOGICAL PRACTICE
Authors Agata Forkasiewicz1, Maja Dorociak1, Kamilla Stach2, Piotr Szelachowski1, Renata Tabola3 and Katarzyna Augoff1*
Abstract One of the hallmarks of cancer cells is increased energy requirements associated with the higher rate of cellular proliferative activity. Metabolic changes in rapidly dividing cancer cells are closely associated with increased uptake of glucose and abnormal activity of lactate dehydrogenase (LDH), which regulates the processing of glucose to lactic acid. As serum LDH levels were found to be commonly increased in cancer patients and correlated with poor clinical outcome and resistance to therapy, the determination of LDH has become a standard supportive tool in diagnosing cancers or monitoring the effects of cancer treatment. The aim of this review is to summarize the current knowledge about methods and the practical utility for measuring both the total LDH and LDH isoenzymatic activities in the diagnosis, prognosis and prediction of cancer diseases.
Keywords Lactate dehydrogenase, LDH isoforms, Warburg effect, Tumor markers
Address and Contact Information 1 Department of Surgical Education, Wroclaw Medical University, ul.Sklodowskiej-Curie 66, 50-369 Wroclaw, Poland
2 Department of Biochemistry, Wroclaw Medical University, Wroclaw, Poland.
3 Second Department and Clinic of General and Oncological Surgery, Wroclaw Medical University, Wroclaw, Poland.
* Corresponding author: katarzyna.augoff@umed.wroc.pl
Read full article at BMC

No. 31DOI: 10.1186/s11658-020-00229-6 Volume 25 (2020) - 25:36
Title SILIBININ SUPPRESSES EPITHELIAL–MESENCHYMAL TRANSITION IN HUMAN NON-SMALL CELL LUNG CANCER CELLS BY RESTRAINING RHBDD1
Authors Suyan Xu, Hongyan Zhang, Aifeng Wang, Yongcheng Ma, Yuan Gan and Guofeng Li*
Abstract Background: Rhomboid domain containing 1 (RHBDD1) plays a crucial role in tumorigenesis. Silibinin, which is a natural extract from milk thistle, has shown anti-tumor effects against various tumors. Here, we investigate whether silibinin affects the function of RHBDD1 in non-small cell lung cancer (NSCLC) cell proliferation, migration and invasion.
Methods: The Oncomine database and an immunohistochemistry (IHC) assay were used to determine the RHBDD1 expression levels in lung cancer tissues. The associations between RHBDD1 and overall survival rate or clinicopathological parameters were respectively assessed using the Kaplan-Meier overall survival analysis or Chi-squared test. CCK-8 and Transwell assays were applied to analyze cell proliferation, migration and invasion. A549 cells were incubated with increasing concentrations of silibinin. RHBDD1 knockdown and overexpression were achieved via transfection with si-RHBDD1 or RHBDD1 overexpression plasmid, respectively. Western blotting was performed to measure the expressions of epithelial–mesenchymal transition (EMT) markers.
Results: We found that overexpression of RHBDD1 in lung cancer tissues correlates with a poor prognosis of survival. Clinical specimen analysis showed that upregulation of RHBDD1 correlates remarkably well with TNM stage and lymph node metastasis. Silibinin suppresses A549 cell proliferation, migration, invasion and EMT in a dose-dependent manner. Importantly, RHBDD1 was downregulated in silibinin-treated A549 cells. RHBDD1 overexpression reversed the suppressive effects of silibinin on A549 cell proliferation, migration, invasion and EMT expression, while its knockdown enhanced them.
Conclusions: These findings shown an anti-tumor impact of silibinin on NSCLC cells via repression of RHBDD1.
Keywords NSCLC, Silibinin, RHBDD1, Epithelial–mesenchymal transition
Address and Contact Information Department of Pharmacy, Henan Provincial People Hospital, Department of Pharmacy of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China
* Corresponding author: guofeng_li245@163.com
Read full article at BMC

No. 32DOI: 10.1186/s11658-020-00224-x Volume 25 (2020) - 25:37
Title Do ABC transporters regulate plasma membrane organization?
Authors Ambroise Wu1†, Karolina Wojtowicz1†, Stephane Savary2, Yannick Hamon3* and Tomasz Trombik1*
Abstract The plasma membrane (PM) spatiotemporal organization is one of the major factors controlling cell signaling and whole-cell homeostasis. The PM lipids, including cholesterol, determine the physicochemical properties of the membrane bilayer and thus play a crucial role in all membrane-dependent cellular processes. It is known that lipid content and distribution in the PM are not random, and their transversal and lateral organization is highly controlled. Mainly sphingolipid- and cholesterol-rich lipid nanodomains, historically referred to as rafts, are extremely dynamic “hot spots” of the PM controlling the function of many cell surface proteins and receptors. In the first part of this review, we will focus on the recent advances of PM investigation and the current PM concept. In the second part, we will discuss the importance of several classes of ABC transporters whose substrates are lipids for the PM organization and dynamics. Finally, we will briefly present the significance of lipid ABC transporters for immune responses.
Keywords ABC transporter, Plasma membrane, Cholesterol, Phospholipids, Rafts, Membrane (nano)domains
Address and Contact Information 1 Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
2 Lab. Bio-PeroxIL EA7270, University of Bourgogne Franche-Comté, Dijon, France.
3 Aix Marseille University, CNRS, INSERM, CIML, Centre d’Immunologie de Marseille-Luminy, Marseille, France
* Corresponding author: hamon@ciml.univ-mrs.fr; tomasz.trombik@uwr.edu.pl
Ambroise Wu and Karolina Wojtowicz contributed equally to this work.
Read full article at BMC

No. 33DOI: 10.1186/s11658-020-00230-z Volume 25 (2020) - 25:38
Title EFFECTS OF HPV16 E6 PROTEIN ON Daxx-INDUCED APOPTOSIS IN C33A CELLS
Authors Shuangyang Tang, Shuang Ding, Lan Yu, Haiyan Shen, Yanping Wan* and Yimou Wu*
Abstract Aims: Daxx is a highly conserved nuclear protein with an important role in transcription, apoptosis and other cell processes. We investigated the role of HPV16 E6 in Daxx-induced apoptosis through their interactions in C33A cells.
Methods: The binding of HPV16 E6 and Daxx was confirmed in C33A cells using co-immunoprecipitation and indirect immunofluorescence assays. Quantitative PCR and western blotting were performed to determine the RNA and protein expressions of Daxx, respectively. Automatic cell count and MTT assays were performed to investigate the proliferation of C33A cells. The apoptosis rate of C33A cells was determined via flow cytometry using Annexin V-FITC/PI staining. The relative activity of caspase-8 was tested using ELISA.
Results: HPV16 E6 can bind with Daxx and cause its translocation in C33A cells. The transfected HPV16 E6 can cause a decrease in relative quantification for Daxx in Daxx-overexpressing cells. After Daxx transfection, cell proliferation was found to decrease sharply and cell apoptosis to increase sharply. However, when HPV16 E6 was co-transfected with Daxx, this decrease and increase both became gentle. Similarly, HPV16 E6 made the Daxx-induced increase in caspase-8 activity milder.
Conclusions: HPV16 E6 is involved in inhibiting apoptosis through deregulation of Daxx-induced caspase-8 activities.
Keywords Human papillomavirus type16, E6 protein, Death domain associated protein, Proliferation, Apoptosis, Caspase-8
Address and Contact Information Pathogenic Biology Institute, University of South China, Hengyang 421001, P. R. China
* Corresponding author: wanyy1991@aliyun.com; wuyimou@126.com
Read full article at BMC

No. 34DOI: 10.1186/s11658-020-00231-y Volume 25 (2020) - 25:39
Title SPECIFIC FUNCTIONS OF EXOSTOSIN-LIKE 3 (EXTL3) GENE PRODUCTS
Authors Shuhei Yamada
Abstract Exostosin-like 3(EXTL3) encodes the glycosyltransferases responsible for the biosynthesis of the backbone structure of heparan sulfate (HS), a sulfated polysaccharide that is ubiquitously distributed on the animal cell surface and in the extracellular matrix. A lack of EXTL3 reduces HS levels and causes embryonic lethality, indicating its indispensable role in the biosynthesis of HS. EXTL3 has also been identified as a receptor molecule for regenerating islet-derived (REG) protein ligands, which have been shown to stimulate islet β-cell growth. REG proteins also play roles in keratinocyte proliferation and/or differentiation, tissue regeneration and immune defenses in the gut as well as neurite outgrowth in the central nervous system. Compared with the established function of EXTL3 as a glycosyltransferase in HS biosynthesis, the REG-receptor function of EXTL3 is not conclusive. Genetic diseases caused by biallelic mutations in the EXTL3 gene were recently reported to result in a neuro-immuno-skeletal dysplasia syndrome. EXTL3 is a key molecule for the biosynthesis of HS and may be involved in the signal transduction of REG proteins.
Keywords Exostosin-like 3 (EXTL3), Heparan sulfate (HS), Biosynthesis, Glycosaminoglycan, Regenerating islet-derived (REG) protein
Address and Contact Information Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan
Corresponding author: shuheiy@meijo-u.ac.jp
Read full article at BMC

No. 35DOI: 10.1186/s11658-020-00232-x Volume 25 (2020) - 25:40
Title FORCED EXPRESSION OF miR-143 AND -145 IN CARDIOMYOCYTES INDUCES CARDIOMYOPATHY WITH A REDUCTIVE REDOX SHIFT
Authors Kota Ogawa1, Akiko Noda1, Jun Ueda2,3†, Takehiro Ogata4†, Rumiko Matsuyama1, Yuji Nishizawa1, Shanlou Qiao1, Satoru Iwata1,2,5, Morihiro Ito1, Yoshitaka Fujihara6,7, Masatoshi Ichihara1, Koichi Adachi8, Yuji Takaoka1 and Takashi Iwamoto1,2*
Abstract Background: Animal model studies show that reductive stress is involved in cardiomyopathy and myopathy, but the exact physiological relevance remains unknown. In addition, the microRNAs miR-143 and miR-145 have been shown to be upregulated in cardiac diseases, but the underlying mechanisms associated with these regulators have yet to be explored.
Methods: We developed transgenic mouse lines expressing exogenous miR-143 and miR-145 under the control of the alpha-myosin heavy chain (αMHC) promoter/enhancer.
Results: The two transgenic lines showed dilated cardiomyopathy-like characteristics and early lethality with markedly increased expression of miR-143. The expression of hexokinase 2 (HK2), a cardioprotective gene that is a target of miR-143, was strongly suppressed in the transgenic hearts, but the in vitro HK activity and adenosine triphosphate (ATP) content were comparable to those observed in wild-type mice. In addition, transgenic complementation of HK2 expression did not reduce mortality rates. Although HK2 is crucial for the pentose phosphate pathway (PPP) and glycolysis, the ratio of reduced glutathione (GSH) to oxidized glutathione (GSSG) was unexpectedly higher in the hearts of transgenic mice. The expression of gamma-glutamylcysteine synthetase heavy subunit (γ-GCSc) and the in vitro activity of glutathione reductase (GR) were also higher, suggesting that the recycling of GSH and its de novo biosynthesis were augmented in transgenic hearts. Furthermore, the expression levels of glucose-6-phosphate dehydrogenase (G6PD, a rate-limiting enzyme for the PPP) and p62/SQSTM1 (a potent inducer of glycolysis and glutathione production) were elevated, while p62/SQSTM1 was upregulated at the mRNA level rather than as a result of autophagy inhibition. Consistent with this observation, nuclear factor erythroid-2 related factor 2 (Nrf2), Jun N-terminal kinase (JNK) and inositol-requiring enzyme 1 alpha (IRE1α) were activated, all of which are known to induce p62/SQSTM1 expression.
Conclusions: Overexpression of miR-143 and miR-145 leads to a unique dilated cardiomyopathy phenotype with a reductive redox shift despite marked downregulation of HK2 expression. Reductive stress may be involved in a wider range of cardiomyopathies than previously thought.
Keywords Reductive stress, microRNA, Cardiomyopathy, G6PD, p62/SQSTM1, JNK, IRE1α
Address and Contact Information 1 Department of Biomedical Sciences, Chubu University Graduate School of Life and Health Sciences, Kasugai, Aichi, Japan.
2 Center for Education in Laboratory Animal Research, Chubu University, Kasugai, Aichi, Japan.
3 Present address: Center for Advanced Research and Education, Asahikawa Medical University, Asahikawa, Hokkaido, Japan.
4 Department of Pathology and Cell Regulation, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Ogawa et al. Cellular & Molecular Biology Letters (2020) 25:40 Page 19 of 21 Kyoto, Japan.
5 College of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi, Japan.
6 Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
7 Present address: Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center, Osaka, Japan.
8 Radioisotope Research Center Medical Division, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.
* Corresponding author: iwamoto@isc.chubu.ac.jp
Jun Ueda and Takehiro Ogata contributed equally to this work.
Read full article at BMC

36
No. DOI: 10.1186/s11658-020-00233-w Volume 25 (2020) - 25:41
Title THE MDM2 LIGAND Nutlin-3 DIFFERENTIALLY ALTERS EXPRESSION OF THE IMMUNE BLOCKADE RECEPTORS PD-L1 AND CD276
Authors Ruidong Li1,2†, Pavlina Zatloukalova3†, Petr Muller3, Maria Gil-Mir1, Sachin Kote4, Simon Wilkinson1, Alain J. Kemp1, Lenka Hernychova3, Yaxin Wang5, Kathryn L. Ball1, Kaixiong Tao2*, Ted Hupp1,3,4* and Borivoj Vojtesek3*
Abstract Background: The links between the p53/MDM2 pathway and the expression of pro-oncogenic immune inhibitory receptors in tumor cells are undefined. In this report, we evaluate whether there is p53 and/or MDM2 dependence in the expression of two key immune receptors, CD276 and PD-L1.
Methods: Proximity ligation assays were used to quantify protein-protein interactions in situ in response to Nutlin-3. A panel of p53-null melanoma cells was created using CRISPR-Cas9 guide RNA mediated genetic ablation. Flow cytometric analyses were used to assess the impact of TP53 or ATG5 gene ablation, as well as the effects of Nutlin-3 and an ATM inhibitor on cell surface PD-L1 and CD276. Targeted siRNA was used to deplete CD276 to assess changes in cell cycle parameters by flow cytometry. A T-cell proliferation assay was used to assess activity of CD4+ T-cells as a function of ATG5 genotype.
Results: CD276 forms protein-protein interactions with MDM2 in response to Nutlin-3, similar to the known MDM2 interactors p53 and HSP70. Isogenic HCT116 p53-wt/null cancer cells demonstrated that CD276 is induced on the cell surface by Nutlin-3 in a p53-dependent manner. PD-L1 was also unexpectedly induced by Nutlin-3, but PD-L1 does not bind MDM2. The ATM inhibitor KU55993 reduced the levels of PD-L1 under conditions where Nutlin-3 induces PD-L1, indicating that MDM2 and ATM have opposing effects on PD-L1 steady-state levels. PD-L1 is also up-regulated in response to genetic ablation of TP53 in A375 melanoma cell clones under conditions in which CD276 remains unaffected. A549 cells with a deletion in the ATG5 gene up-regulated only PD-L1, further indicating that PD-L1 and CD276 are under distinct genetic control.
Conclusion: Genetic inactivation of TP53, or the use of the MDM2 ligand Nutlin-3, alters the expression of the immune blockade receptors PD-L1 and CD276. The biological function of elevated CD276 is to promote altered cell cycle progression in response to Nutlin-3, whilst the major effect of elevated PD-L1 is T-cell suppression. These data indicate that TP53 gene status, ATM and MDM2 influence PD-L1 and CD276 paralogs on the cell surface. These data have implications for the use of drugs that target the p53 pathway as modifiers of immune checkpoint receptor expression.
Keywords MDM2, p53, Gene editing, Protein-protein interactions, Nutlin-3
Address and Contact Information 1 University of Edinburgh, Institute of Genetics and Molecular Medicine, Edinburgh, Scotland EH4 2XR, UK
2 Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
3 RECAMO, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
4 University of Gdansk, International Centre for Cancer Vaccine Science, ul. Wita Stwosza 63, 80-308 Gdansk, Poland.
5 Department of Anesthesiology and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.

* Corresponding author: kaixiongtao@hust.edu.cn; ted.hupp@ed.ac.uk; vojtesek@mou.cz
Ruidong Li and Pavlina Zatloukalova contributed equally to this work.
Read full article at BMC

No. 37DOI: 10.1186/s11658-020-00234-9 Volume 25 (2020) - 25:42
Title COMPARATIVE TRANSCRIPTOME ANALYSIS OF THE MAIN BEAM AND BROW TINE OF SIKA DEER ANTLER PROVIDES INSIGHTS INTO THE MOLECULAR CONTROL OF RAPID ANTLER GROWTH
Authors Baojin Yao1†, Chaonan Wang2†, Zhenwei Zhou1, Mei Zhang3, Daqing Zhao1, Xueyuan Bai1* and Xiangyang Leng4*
Abstract Background: Deer antlers have become a valuable model for biomedical research due to the capacities of regeneration and rapid growth. However, the molecular mechanism of rapid antler growth remains to be elucidated. The aim of the present study was to compare and explore the molecular control exerted by the main beam and brow tine during rapid antler growth.
Methods: The main beams and brow tines of sika deer antlers were collected from Chinese sika deer (Cervus nippon) at the rapid growth stage. Comparative transcriptome analysis was conducted using RNA-Seq technology. Differential expression was assessed using the DEGseq package. Functional Gene Ontology (GO) enrichment analysis was accomplished using a rigorous algorithm according to the GO Term Finder tool, and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway enrichment analysis was accomplished with the R function phyper, followed by the hypergeometric test and Bonferroni correction. Quantitative real-time polymerase chain reaction (qRT-PCR) was carried out to verify the RNA levels for differentially expressed mRNAs.
Results: The expression levels of 16 differentially expressed genes (DEGs) involved in chondrogenesis and cartilage development were identified as significantly upregulated in the main beams, including transcription factor SOX-9 (Sox9), collagen alpha-1(II) chain (Col2a1), aggrecan core protein (Acan), etc. However, the expression levels of 17 DEGs involved in endochondral ossification and bone formation were identified as significantly upregulated in the brow tines, including collagen alpha-1(X) chain (Col10a1), osteopontin (Spp1) and bone sialoprotein 2 (Ibsp), etc.
Conclusion: These results suggest that the antler main beam has stronger growth capacity involved in chondrogenesis and cartilage development compared to the brow tine during rapid antler growth, which is mainly achieved through regulation of Sox9 and its target genes, whereas the antler brow tine has stronger capacities of endochondral bone formation and resorption compared to the main beam during rapid antler growth, which is mainly achieved through the genes involved in regulating osteoblast and osteoclast activities. Thus, the current research has deeply expanded our understanding of the intrinsic molecular regulation displayed by the main beam and brow tine during rapid antler growth.
Keywords Deer antler, Main beam, Brow tine, RNA-Seq, Molecular mechanism
Address and Contact Information 1 Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
2 College of traditional Chinese medicine, Changchun University of Chinese Medicine, Changchun 130117, China.
3 Innovation Practice Center, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
4 The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
* Correspondence: 23165956@qq.com; leng_xiangyang@163.com
† Baojin Yao and Chaonan Wang contributed equally to this work.
Read full article at BMC

No. 38DOI: 10.1186/s11658-020-00235-8 Volume 25 (2020) - 25:43
Title KNOCKDOWN OF LINC00665 INHIBITS PROLIFERATION AND INVASION OF BREAST CANCER VIA COMPETITIVE BINDING OF miR-3619-5p AND INHIBITION OF CATENIN BETA 1
Authors Minhao Lv1, Qixin Mao1, Juntao Li1, Jianghua Qiao1, Xiuchun Chen1 and Suxia Luo2*
Abstract Background: Long intergenic non-protein coding RNA00665 (LINC00665) plays a crucial tumorigenic role in many cancers, such as gastric cancer and lung adenocarcinoma. However, its role and mechanism of action in the progression of breast cancer (BC) are unknown.
Methods: LINC00665 expression levels were determined using quantitative polymerase chain reaction analysis with BC tissues and cell lines. BC cell proliferation was tested by performing 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays, whereas BC cell migration and invasion capabilities were analyzed by performing transwell migration assays. Percentages of apoptotic cells were measured by flow cytometry. Interactions between LINC00665 and miR-3169-5p were examined by performing luciferase reporter assays, and the expression levels of proteins, such as β-catenin, were examined by western blot analysis.
Results: LINC00665 was expressed at high levels in BC tissues and cells. Upregulated LINC00665 expression correlated with tumor size and tumor, node, and metastasis stages, but not with the age of patients. LINC00665 knockdown inhibited BC cell proliferation, migration, and invasion, whereas it promoted apoptosis. Moreover, bioinformatics analysis and the luciferase reporter assay revealed that LINC00665 bound the microRNA (miR) miR-3619-5p. miR-3619-5p expression correlated negatively with LINC00665 expression in BC tissues. miR-3619-5p overexpression inhibited BC cell proliferation, migration, and invasion, but promoted apoptosis. Simultaneous knockdown of LINC00665 and miR-3619-5p led to increased cell proliferation, migration, and invasion, and inhibited apoptosis. Additionally, catenin beta 1, which encodes the β-catenin protein, was the target gene of miR-3619-5p. β-catenin expression clearly decreased after LINC00665 knockdown and miR-3619-5p overexpression, but increased after simultaneous knockdown of LINC00665 and miR-3619-5p.
Conclusion: LINC00665 knockdown inhibited BC cell proliferation and invasion by binding miR-3619-5p and inhibiting β-catenin expression.
Keywords β-Catenin, Breast cancer, CTNNB1, LINC00665, miR-3619-5p
Address and Contact Information 1 Department of Breast Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China.
2 Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, No. 127, Dongming Road, Jinshui District, Zhengzhou 450008, Henan, P.R. China
* Correspondence: luosxrm@sina.com
Read full article at BMC

No. 39DOI: 10.1186/s11658-020-00236-7 Volume 25 (2020) - 25:44
Title ARSENIC TRIOXIDE AS A NOVEL ANTI‐GLIOMA DRUG: A REVIEW
Authors Yi Fang and Zhen Zhang*
Abstract Arsenic trioxide has shown a strong anti-tumor efect with little toxicity when used in the treatment of acute promyelocytic leukemia (APL). An efect on glioma has also been shown. Its mechanisms include regulation of apoptosis and autophagy; pro-motion of the intracellular production of reactive oxygen species, causing oxidative damage; and inhibition of tumor stem cells. However, glioma cells and tissues from other sources show diferent responses to arsenic trioxide. Researchers are working to enhance its efcacy in anti-glioma treatments and reducing any adverse reactions. Here, we review recent research on the efcacy and mechanisms of action of arsenic trioxide in the treatment of gliomas to provide guidance for future studies.
Keywords Arsenic trioxide, Glioma, Anti-cancer mechanism
Address and Contact Information Department of Ultrasound, First Afliated Hospital of China Medical University, Shenyang 110001, Liaoning, People’s Republic of China
*Corresponding author: 2662898158@qq.com
Read full article at BMC

No. 40DOI: 10.1186/s11658-020-00237-6 Volume 25 (2020) - 25:45
Title UPREGULATION OF miR‐361‐3p SUPPRESSES SEROTONIN‐INDUCED PROLIFERATION IN HUMAN PULMONARY ARTERY SMOOTH MUSCLE CELLS BY TARGETING SERT
Authors Ying Zhang1†, Yongbin Chen2†, Guo Chen1, Yingling Zhou1, Hua Yao1* and Hong Tan1*
Abstract Background: Abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs) is a key mechanism in pulmonary arterial hypertension (PAH). Serotonin (5-hydroxytryptamine, 5-HT) can induce abnormal proliferation of PASMCs. The role of miR-361-3p in serotonin-induced abnormal PASMCs proliferation remains unclear.
Methods: The miR-361-3p level was analyzed in plasma from PAH patients and normal controls and in human PASMCs (hPASMCs) using RT-PCR. The hPASMCs were transfected with an miR-361-3p mimic and then treated with serotonin. Untransfected hPASMCs were used as the control. Cell proliferation was evaluated using an MTS assay and 5-ethynyl-2′-deoxyuridine (EdU) staining. The cell cycle stages were evaluated using fow cytometry. The association between miR-361-3p and serotonin transporter (SERT) was determined using a luciferase reporter assay and anti-AGO2 RNA immunoprecipitation assay. The protein expression was evaluated via western blotting.
Results: The miR-361-3p level was lower in plasma from PAH patients than in plasma from the any of the normal control subjects. The mean pulmonary arterial pressure, pulmonary vascular resistance and pulmonary vascular resistance index were higher in PAH patients whose miR-361-3p level was lower than the median value for patients than in those whose miR-361-3p level was higher than the median. Serotonin treatment reduced miR-361-3p expression in the hPASMCs. MiR-361-3p overexpression suppressed cell proliferation, promoted apoptosis, induced G1 arrest, and decreased the phosphorylation level of ERK1/2 in serotonin-treated hPASMCs. SERT was identifedas an miR-361-3p target. Its overexpression alleviated the efect of miR-361-3p overexpression on serotonin-induced hPASMC proliferation and upregulation of phosphorylated ERK1/2.
Conclusions: The miR-361-3p level is lower in the plasma of PAH patients. Upregulation of miR-361-3p suppresses serotonin-induced proliferation of hPASMCs by targeting SERT. Our results suggest that miR-361-3p is a potential therapeutic target in PAH.
Keywords Abnormal proliferation, Pulmonary artery smooth muscle cells, Serotonin, miR-361-3p
Address and Contact Information 1 Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong 510080 Guangzhou, P. R. China
2 Department of Cardiac Surgery, Guang-dong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 510080 Guangzhou, P. R. China.
* Corresponding author: yaohua2078@163.com; tanhong12345@126.com
Ying Zhang and Yongbin Chen contributed equally to this work.
Read full article at BMC

No. 41DOI: 10.1186/s11658-020-00239-4 Volume 25 (2020) - 25:46
Title OBITUARY: IN MEMORY OF PROFESSOR TADEUSZ CHOJNACKI (1932–2020)
Authors Ewa Swiezewska1* and Anna Chojnacka2
Abstract Professor Tadeusz Chojnacki passed away on June 13, 2020.
Professor Chojnacki was a full member of the Polish Academy of Sciences (PAS), a co-founder and vice-director of the Institute of Biochemistry and Biophysics (IBB) of the Polish Academy of Sciences, and a member of the Scientifc Council of IBB PAS and the Scientifc Councils of a number of scientifc institutions.
Professor Chojnacki was also an organizer of the scientifc life in Poland, serving as the Secretary and Chairman of the Second Department of Biological Sciences of the Polish Academy of Sciences and the Chairman of the Committee of Biochemistry and Biophysics PAS. He was also among the founders of the journal Cellular and Molecular Biology Letters and a member of its Editorial Board.
Professor Chojnacki obtained numerous awards and decorations, including the Commander’s Cross of the Order of Polonia Restituta (2008), the Doctor Honoris Causa Award of the Karolinska Institutet in Stockholm (1986), and the Michał Oczapowski Medal (2015).
Professor Chojnacki graduated from the Medical Department of Warsaw Medical Academy in 1955, and a few years after working as a general practitioner he started his professional carrier as a biochemist. From 1958 he was employed at the Institute of Biochemistry and Biophysics PAS in Warsaw, where he received his PhD (1961), headed the Department of Phospholipids/Department of Lipid Biochemistry at IBB PAS (until 2003) and led numerous research projects.
Professor Chojnacki devoted his entire scientifc career to lipids. Initially, Professor Chojnacki’s projects were focused on phospholipid metabolism (PhD dissertation and a postdoctoral project performed with Professor Gordon Ansell, University of Birmingham with two Nature papers in 1962 and 1966), nucleotides and glycolipids (collaboration with Professor Tadeusz Korzybski - a doyen of Polish biochemistry), as well as the use of isotope methods in biochemistry and laboratory diagnostics (diagnostics of galactosemia, 1969).
Subsequently, Professor Chojnacki initiated pioneer studies on polyisoprenoids, compounds found in all kingdoms of life, and became a world-recognized authority in this feld. Besides studies of polyisoprenoid chemical structure, biosynthesis and biochemistry, he was especially fascinated by chemotaxonomic aspects of plant polyisoprenoids, and their occurrence in various habitats all over the globe. Due to this successfully developed project, IBB PAS became a reference center ofering isoprenoid compounds worldwide through the Collection of Polyprenols established by him. Until very recently, Professor Chojnacki remained scientifcally active, and the most recent topic which attracted his attention was the use of semisynthetic polyisoprenoid derivatives as lipofectants.
Professor Tadeusz Chojnacki established and efectively conducted numerous collaborative projects with research groups from Poland and internationally. Tese included projects spanning more than 35 years (1978–2015) together with Professor Gustav Dallner and Professor Kerstin Brismar, University of Stockholm and Karolinska Institutet, resulting in 46 joint publications. Several projects were performed in collaboration with researchers from the Zielinski Institute of Organic Chemistry, Russian Academy of Sciences in Moscow (Professor Vladimir Shibaev, Dr Tatiana Druzhinina and Dr. Leonid Danilov, who incidentally passed away the same day, June 13, 2020), from the Institute of Chemistry, Academy of Sciences of Moldova in Kishinev (Professors Pavel Vlad and Veaceslaw Kulcitki), from Tohoku University in Sendai (Professor Hiroshi Sagami), and many others.
Tere were also numerous Polish research groups involved in collaboration with Professor Chojnacki, including the Faculty of Biochemistry, Biophysics and Biotechnology Jagiellonian University (Professors Włodzimierz Korohoda, Zbigniew Madeja, Kazimierz Strzałka, Dr. Monika Rak), the Institute of Organic Chemistry PAS (Professors Marek Chmielewski, Jan Pyrek, Włodzimierz Daniewski, Dr. Marek Masnyk), and the Centre of Molecular and Macromolecular Studies PAS in Łódź (Professors Barbara Nawrot and Wojciech Stec). In parallel, Professor Chojnacki’s fascination with plant chemotaxonomy resulted in close collaboration with botanists from numerous botanical gardens and arboreta, including the Botanical Garden—Center for Conservation of Biological Diversity PAS in Powsin (Dr. Andrzej Marczewski, Professors Jerzy Puchalski and Wojciech Dmuchowski), the Arboretum in Trivandrum and in Lucknow, the Arboretum in Bolestraszyce, and the Institute of Dendrology in Kornik. Professor Chojnacki published approximately 200 papers, many of them considered as landmarks in the feld, and he supervised 9 PhD dissertations. Professor Tadeusz Chojnacki was a great scientist, a teacher and mentor who shared his knowledge and skills with us—his students and collaborators.
We will miss his comments on life and science, his advice and suggestions. And we will miss him.
Address and Contact Information 1 Department of Lipid Biochemistry, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5A, 02‐106 Warsaw, Poland.
2 Department of the History and Theory of Theatre, Institute of Art, Polish Academy of Sciences, Długa 26/28, 00‐238 Warsaw, Poland.
Read full article at BMC

No. 42DOI: 10.1186/s11658-020-00238-5 Volume 25 (2020) - 25:47
Title hsa_circ_0006916 PROMOTES HEPATOCELLULAR CARCINOMA PROGRESSION BY ACTIVATING THE miR‐337‐3p/STAT3 AXIS
Authors Xiao‐Yong Zhou*, Hui Yang, Yang‐Qiu Bai, Xiu‐Ling Li, Shuang‐Yin Han and Bing‐Xi Zhou
Abstract Background: Circular RNAs (circRNAs) are thought to be involved in the development of various malignancies. The expression and function of hsa_circ_0006916, a newly identifed circRNA, in hepatocellular carcinoma remain unclear.
Methods: Quantitative RT-PCR was used to detect hsa_circ_0006916 in hepatocellular carcinoma. In vitro function assays were conducted to explore growth and invasion of hepatocellular carcinoma cells. Next, the mechanism of hsa_circ_0006916 function in hepatocellular carcinoma was determined by luciferase reporter and RIP assays.
Results: Hsa_circ_0006916 was substantially overexpressed in hepatocellular carcinoma tissues and cells. High levels of hsa_circ_0006916 in hepatocellular carcinoma patients were associated with advanced clinical characteristics. Down-regulation of hsa_circ_0006916 decreased the growth and invasion of hepatocellular carcinoma cells in vitro. The results suggested that hsa_circ_0006916 acted as a sponge of miR-337-3p and had an important functional use in the regulation of STAT3 levels in hepatocellular carcinoma cells. Moreover, miR-337-3p inhibition or STAT3 overexpression abolished the efect of hsa_circ_0006916 suppression on the progression of hepatocellular carcinoma cells.
Conclusions: Our data suggest a novel hsa_circ_0006916/miR-337-3p/STAT3 axis in hepatocellular carcinoma, and provide a new target for treatment.
Keywords hsa_circ_0006916, miR-337-3p, STAT3, Hepatocellular carcinoma
Address and Contact Information Department of Gastroenterology and Hepatology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou 450003, Henan, China
*Corresponding author: zhouxiaoyongzxy@163.com
Read full article at BMC

No. 43DOI: 10.1186/s11658-020-00241-w Volume 25 (2020) - 25:48
Title THE NUCLEAR RECEPTOR 4A FAMILY MEMBERS: MEDIATORS IN HUMAN DISEASE AND AUTOPHAGY
Authors Liqun Chen1,2*† , Fengtian Fan1,2†, Lingjuan Wu1,2 and Yiyi Zhao1,2
Abstract The Nuclear receptor 4A (NR4A) subfamily, which belongs to the nuclear receptor (NR) superfamily, has three members: NR4A1 (Nur77), NR4A2 (Nurr1) and NR4A3 (Nor1). They are gene regulators with broad involvement in various signaling pathways and human disease responses, including autophagy. Here, we provide a concise overview of the current understanding of the role of the NR4A subfamily members in human diseases and review the research into their regulation of cell autophagy. A deeper understanding of these mechanisms has potential to improve drug development processes and disease therapy.
Keywords NR4A family, Autophagy, Human disease, Nur77, Nurr1, Nor1
Address and Contact Information 1 College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
2 Institute of Apply Genomics, Fuzhou University, Fuzhou 350108, China.
*Corresponding author: lqchen@fzu.edu.cn
Liqun Chen and Fengtian Fan are co-frst author.
Read full article at BMC

No. 44DOI: 10.1186/s11658-020-00240-x Volume 25 (2020) - 25:49
Title MicroRNA‐296‐5p INHIBITS CELL METASTASIS AND INVASION IN NASOPHARYNGEAL CARCINOMA BY REVERSING TRANSFORMING GROWTH FACTOR‐β‐INDUCED EPITHELIAL–MESENCHYMAL TRANSITION
Authors Meihui Chen1,2†, Chen Chen1†, Haiqing Luo3†, Jing Ren1, Qiuqin Dai1, Wenjia Hu1, Keyuan Zhou1, Xudong Tang1* and Xiangyong Li1*
Abstract Aim: To explore the efect of miR-296-5p on the metastasis of nasopharyngeal carcinoma (NPC) cells and investigate the underlying mechanism.
Methods: The expressions of miR-296-5p in NPC tissues and cells were determined using GSE32920 database analysis and real-time PCR and miRNA microarray assays. An miR-296-5p mimic and inhibitor were transfected into NPC cells. Then, immunofuorescence imaging, scratch wound-healing, transwell migration and invasion assays were used to observe the efects of miR-296-5p on cell metastasis and invasion. Real-time PCR and western blotting were carried out to detect the expressions of genes and proteins related to epithelial–mesenchymal transition (EMT). A dual luciferase reporter assay was used to identify whether TGF-β is the target gene of miR-296-5p. Finally, TGF-β expression plasmids were transfected into NPC cells to verify the role of TGF-β in the miR-296-5p-mediated inhibition of nasopharyngeal carcinoma cell metastasis.
Results: Our results show that miR-296-5p inhibits the migratory and invasive capacities of NPC cells by targeting TGF-β, which suppresses EMT. Importantly, the miR-296-5p level was signifcantly lower in human NPC tissues than in adjacent normal tissues. It also negatively correlated with TGF-β and was signifcantly associated with the lymph node metastasis of patients with NPC.
Conclusions: Our fndings show that miR-296-5p represses the EMT-related metastasis of NPC by targeting TGF-β. This provides new insight into the role of miR-296-5p in regulating NPC metastasis and invasiveness.
Keywords miR-296-5p, Metastasis, Invasion, EMT, Nasopharyngeal carcinoma
Address and Contact Information 1 Institute of Biochemistry and Molecular Biology of Guangdong Medical University, No. 2 Wenming Dong Road, Xiashan District, Zhanjiang 524023, Guangdong, China
2 Department of Clinical Laboratory of Zhanjiang Central Hospital, Zhan-jiang 524023, China. 3 Center of Oncology of The Afliated Hospital of Guangdong Medical University, Zhanjiang 524023, China.
*Corresponding author: tangxudong2599@126.com; xyli75@126.com
Meihui Chen, Chen Chen and Haiqing Luo contributed equally to this manuscript.
Read full article at BMC

No. 45DOI: 10.1186/s11658-020-00242-9 Volume 25 (2020) - 25:50
Title IN VITRO DIFERENTIATION OF W8B2+ HUMAN CARDIAC STEM CELLS: GENE EXPRESSION OF IONIC CHANNELS AND SPONTANEOUS CALCIUM ACTIVITY
Authors Oualid Ayad1, Zeina R. Al Sayed3, Stéphane Sebille1, Christophe Magaud1, Charles‐Albert Chapotte‐Baldacci1, Christophe Jayle2, Jean‐François Faivre1, Nathalie Gaborit3, Aurélien Chatelier1† and Patrick Bois1*†
Abstract Background: Human cardiac stem cells expressing the W8B2 marker (W8B2+ CSCs) were recently identifed and proposed as a new model of multipotent CSCs capable of diferentiating into smooth muscle cells, endothelial cells and immature myocytes. Nevertheless, no characterization of ion channel or calcium activity during the diferentiation of these stem cells has been reported.
Methods: The objectives of this study were thus to analyze (using the TaqMan Low-Density Array technique) the gene profle of W8B2+ CSCs pertaining to the regulation of ion channels, transporters and other players involved in the calcium homeostasis of these cells. We also analyzed spontaneous calcium activity (via the GCaMP calcium probe) during the in vitro diferentiation of W8B2+ CSCs into cardiac myocytes.
Results: Our results show an entirely diferent electrophysiological genomic profle between W8B2+ CSCs before and after diferentiation. Some specifc nodal genes, such as Tbx3, HCN, ICaT, L, KV, and NCX, are overexpressed after this diferentiation. In addition, we reveal spontaneous calcium activity or a calcium clock whose kinetics change during the diferentiation process. A pharmacological study carried out on diferentiated W8B2+ CSCs showed that the NCX exchanger and IP3 stores play a fundamental role in the generation of these calcium oscillations.
Conclusions: Taken together, the present results provide important information on ion channel expression and intrinsic calcium dynamics during the diferentiation process of stem cells expressing the W8B2 marker.
Keywords W8B2+ human cardiac stem cell, Cardiac diferentiation, Calcium activity, Oscillations, Ion channels, GCaMP
Address and Contact Information 1 University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
2 CHU of Poitiers chirurgie cardiaque et thoracique, , Poitiers Cedex 09, France.
3 CNRS, INSERM, l’institut du thorax, Université de Nantes, 44000 Nantes, France.
*Corresponding author: Patrick.Bois@univ‐poitiers.fr
Aurélien Chatelier and Patrick Bois have contributed equally to this work
Read full article at BMC