Monday, February 29, 2016

Smoking-induced expression of the GPR15 gene indicates its potential role in chronic inflammatory pathologies

Significance Statement

Tobacco smoking is considered the leading preventable cause of morbidity and mortality. Smoking affects more than 1 billion individuals worldwide and accounts for an estimated 3 million deaths per year. However, the exact molecular mechanisms how smoking influences the organism, is still not understood. Smoking-induced molecular alterations have been analysed in previous studies and peripheral gene-expression profiling has allowed for the detection of early molecular signatures relevant to the toxic effects of smoking. In addition to the toxic effects, smoking also induces prominent immunological changes in the lungs. The aim of our study was to find systemic changes that are caused by smoking. We performed whole transcriptome analysis of the peripheral blood in smokers and non-smokers. As a results we found significant up-regulation of GPR15, that is chemoattractant and regulates the homing of immune cells. The activation of GPR15 expression was coincided with the hypomethylation of GPR15 locus. Our study indicates that smoking causes systemic molecular changes that regulate innate immunity and chronic inflammation. Our results support the understanding that the smoking health effects are mediated by inflammatory response.

Smoking-induced expression of the GPR15 gene indicates its potential role in chronic inflammatory pathologies. Global medical discovery

Journal Reference

Am J Pathol. 2015;185(11):2898-906.

Kõks G1, Uudelepp ML2, Limbach M1, Peterson P1, Reimann E3, Kõks S4.

Show Affiliations
  1. Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.
  2. Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia; Department of Genetics, Tartu University Hospital, Tartu, Estonia.
  3. Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia; Department of Reproductive Biology, Estonian University of Life Sciences, Tartu, Estonia.
  4. Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia; Department of Reproductive Biology, Estonian University of Life Sciences, Tartu, Estonia. Electronic address: sulev.koks@ut.ee.

Abstract

Despite the described clear epigenetic effects of smoking, the effect of smoking on genome-wide gene expression in the blood is obscure. We therefore studied the smoking-induced changes in the gene-expression profile of the peripheral blood. RNA was extracted from the whole blood of 48 individuals with a detailed smoking history (24 never-smokers, 16 smokers, and 8 ex-smokers). Gene-expression profiles were evaluated with RNA sequencing, and results were analyzed separately in 24 men and 24 women. In the male smokers, 13 genes were statistically significantly (false-discovery rate <0.1) differentially expressed; in female smokers, 5 genes. Although most of the differentially expressed genes were different between the male and female smokers, the G-protein-coupled receptor 15 gene (GPR15) was differentially expressed in both male and female smokers compared with never-smokers. Analysis of GPR15 methylation identified significantly greater hypomethylation in smokers compared with that in never-smokers. GPR15 is the chemoattractant receptor that regulates T-cell migration and immunity. Up-regulation of GPR15 could explain to some extent the health hazards of smoking with regard to chronic  inflammatory diseases.

Copyright © 2015 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.

Go To Am J Pathol

 

 

SLAT promotes TCR-mediated, Rap1-dependent LFA-1 activation and adhesion through interaction of its PH domain with Rap1

SLAT promotes TCR-mediated, Rap1-dependent LFA-1 activation and adhesion through interaction of its PH domain with Rap1

Journal Reference

J Cell Sci. 2015 Dec 1;128(23):4341-52.

Côte M1, Fos C1, Canonigo-Balancio AJ1, Ley K2, Bécart S3, Altman A3.

Show Affiliations
  1. Division of Cell Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA.
  2. Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA.
  3. Division of Cell Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA sbecart@its.jnj.com, amnon@lji.org.

Abstract

SLAT (also known as DEF6) promotes T cell activation and differentiation by regulating NFAT-Ca(2+) signaling. However, its role in TCR-mediated inside-out signaling, which induces integrin activation and T cell adhesion, a central process in T cell immunity and inflammation, has not been explored. Here, we show that SLAT is crucial for TCR-induced adhesion to ICAM-1 and affinity maturation of LFA-1 in CD4(+) T cells. Mechanistic studies revealed that SLAT interacts, through its PH domain, with a key component of inside-out signaling, namely the active form of the small GTPase Rap1 (which has two isoforms, Rap1A and Rap1B). This interaction has been further shown to facilitate the interdependent recruitment of Rap1 and SLAT to the T cell immunological synapse upon TCR engagement. Furthermore, a SLAT mutant lacking its PH domain drastically inhibited LFA-1 activation and CD4(+) T cell adhesion. Finally, we established that a constitutively active form of Rap1, which is present at the plasma membrane, rescues the defective LFA-1 activation and ICAM-1 adhesion in SLAT-deficient (Def6(-/-)) T cells. These findings ascribe a new function to SLAT, and identify Rap1 as a target of SLAT function in TCR-mediated inside-out signaling.

© 2015. Published by The Company of Biologists Ltd.

Go To J Cell Sci

Structural Insight into the Mechanism of TFIIH Recognition by the Acidic String of the Nucleotide Excision Repair Factor XPC

Significance Statement

The genome is constantly confronted with the risk of DNA damage by various factors such as hazardous metabolic byproducts, carcinogens, and UV radiation. Nucleotide excision repair (NER) is one of DNA repair systems that organisms have evolved to protect the genome. Dysfunction of the NER pathway causes rare genetic disorders; xeroderma pigmentosum, Cockayne syndrome, and trichothiodystrophy. In mammalian global genomic NER, XPC (xeroderma pigmentosum group C complementing protein) detects DNA lesions and then recruits a ten-subunit complex, TFIIH to the lesion to open up the damaged DNA. Although TFIIH is mostly engaged in transcription as a general transcription factor under normal culture conditions, it rapidly switches to sites of DNA damage upon UV irradiation of cells. However, the precise mechanism underlying the recruitment of TFIIH to DNA lesions by XPC has remained unclear. In the present study, Okuda et al. have determined the structure of the acidic region of human XPC bound to the pleckstrin homology (PH) domain of TFIIH p62 using nuclear magnetic resonance (NMR) spectroscopy. XPC uses a coupled folding and binding mode, and wraps around the basic surface of the p62 PH domain. The bound structure of XPC closely resembles the extended acidic string-like structures observed for the general transcription factor TFIIEα and the tumor suppressor p53 bound to the p62 PH domain; however, the structure reveals critical differences in the recognition site. The key residues of XPC necessary for strong binding have been verified by mutational analyses using isothermal titration calorimetry (ITC) in vitro and immunoprecipitation in vivo. Alanine substitution of these key residues compromised the recruitment of TFIIH to sites of DNA damage, UV resistance, and the repair of UV-induced pyrimidine-pyrimidone (6-4) photoproducts in XPC-deficient cells stably transformed to express XPC protein. This study sheds light on the mechanism for functional cooperation between XPC and TFIIH in the early stage of global genomic NER.

 

About The Author

Dr. Masahiko Okuda received his PhD degree in 2000 from Yokohama City University in Japan, working on the structural studies of the general transcription factor TFIIEβ with Professor Yoshifumi Nishimura. He also worked with Prof. Nishimura as a postdoctoral fellow. He obtained a contract assistant professor position in 2008. His main research interest is in the structural basis of DNA repair and gene regulation in eukaryotes.  

 

About The Author

Prof. Yoshifumi Nishimura received his Ph. D degree in 1976 from Faculty of Pharmaceutical Sciences of the University of Tokyo on UV resonance Raman spectroscopy of biological molecules, and then worked as an instructor and Associate Professor in the Faculty. In 1989, he moved into Graduate School of Yokohama City University as a Professor and worked on NMR spectroscopy of several transcription factors. Professor Nishimura has been working on structural epi-genomics by NMR, and now is an Adviser to the President of Yokohama City University and also a Project Leader of NMR Platform, which contains 950 MHz, 800 MHz, 700 MHz, 600 MHz, and 500 MHz NMR spectrometers, in the Graduate School of Medical Life Science of Yokohama City University.  

Figure Legend:  Solution structure of the complex formed between the XPC acidic string and the TFIIH p62 PH domain. Overlay of the backbone structures of the 20 best structures (ribbon representation). XPC is shown in rainbow color and TFIIH p62 in light blue. 

Structural Insight into Mechanism of TFIIH Recognition by Acidic String of Nucleotide Excision Repair Factor XPC. Global medical discovery

Journal Reference

Structure. 2015;23(10):1827-37. 

Okuda M1, Kinoshita M2, Kakumu E2, Sugasawa K2, Nishimura Y3.

Show Affiliations
  1. Graduate School of Medical Life Science, Yokohama City University, 1-7-29, Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
  1. Biosignal Research Center, Organization of Advanced Science and Technology, 1-1,    Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan; Department of Biology, Graduate,    School of Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501,  Japan.
  1. Graduate School of Medical Life Science, Yokohama City University, 1-7-29, Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan; nisimura@tsurumi.yokohama-cu.ac.jp.

Abstract

In global genome repair (GGR), XPC detects damaged nucleotides and recruits TFIIH complex. The small acidic region of XPC binds to the pleckstrin homology (PH) domain of TFIIH subunit p62; however, the recognition mechanism remains elusive. Here, we use nuclear magnetic resonance to present the tertiary structure of XPC bound to the PH domain. The XPC acidic region forms a long string stabilized by insertion of Trp133 and Val136 into two separate hollows of the PH domain, coupled with extensive electrostatic contacts. Analysis of several XPC mutants revealed that particularly Trp133 is essential for binding to the PH domain. In cell lines stably expressing mutant XPC, alanine substitution at Trp133 or Trp133/Val136 compromised UV resistance, recruitment of TFIIH to DNA damage, and removal of UV-induced photoproducts from genomic DNA. These findings show how TFIIH complex is recruited by XPC to damaged DNA, advancing our understanding of the early stage of GGR.

Copyright © 2015 Elsevier Ltd. All rights reserved.

Go To Structure

 

 

Differential Reovirus-Specific and Herpesvirus-Specific Activator Protein 1 Activation of Secretogranin II Leads to Altered Virus Secretion

Significance Statement

There has been considerable work performed studying viruses and how they use hosts to replicate. However, much remains unknown about virus-host interactions and how they may be virus-specific.  In a previous quantitative study, we found reovirus strain T1L induced upregulation of the host secretogranin II (SCG2) protein, a protein not previously examined with respect to virus infections. The current study aimed to determine whether secretogranin II plays a role in infection. Herpes simplex type 1 virus also was studied because it is a ubiquitous virus that has co-evolved with humans over a very long time and is able to evade the host immune system and establish latent infections. We found that reovirus-induced secretogranin II upregulation, and phosphorylation, began at 18 hours post infection. Conversely, HSV-1 infection led to secretogranin II down regulation as early as 6 hours post infection. There was a negative correlation in the amount of secretogranin II and release of infectious virus from the cell. Analyses of the host AP-1 transcription pathway, known to activate secretogranin II expression, revealed a virus-specific difference in the secretogranin II activation pathway. This work aids in furthering our knowledge of virus-host interactions.

About The Author

Dr. Alicia Berard obtained her B.Sc. honours degree in Biochemistry from the University of Winnipeg, Canada in 2006. She began her graduate studies at the University of Manitoba, Canada under both Dr. Alberto Severini and Dr. Kevin Coombs supervisions. She recently received her PhD, in 2015, in Medical Microbiology where her work focused on host proteomics in reovirus and herpes simplex virus infections. She was supported during her degree with fellowship awards including the Manitoba Graduate Scholarship and Manitoba Health Research Council Health Sciences Centre Studentship awards. She is currently a postdoctoral fellow at the JC Wilt Infectious Diseases Research Centre, Canada, working on host proteomics in SIV/HIV infection in Dr. Adam Burgener’s laboratory.

About The Author

Dr. Kevin Coombs received his B.A.s in Biology and English from the State University of New York in Geneseo, N.Y., and his M.A. and Ph.D degrees from the University of Texas in Austin, TX. His post-doctoral training in molecular and structural virology was done in the labs of Dr. Bernard Fields at Harvard Medical School and Dr. Steven Harrison at Harvard University. Dr. Coombs is presently a Professor in the Department of Medical Microbiology and is Assistant Dean of Research for the College of Medicine in the Faculty of Health Sciences. He serves on several Editorial Boards of journals publishing in Cell Biology, Molecular Biology and Virology, has served as a panel member on NIH and AHFMR Peer Review Committees and as Scientific Officer and panel member on the CIHR Virology and Viral Pathogenesis Committee. His research interests include delineation of the protein and nucleic acid interactions in nucleoprotein complexes, using a variety of RNA viruses as models. This work is in general areas of:

* Generation and molecular characterization of assembly-defective virus mutants

* Inhibition of virus replication using pharmacologic inhibitors

* Mass spectrometry- and Systems-based analyses of virus and host protein alterations

* Development of non-pathogenic viruses as “bio-indicators” for wastewater and medical instrument disinfection.

Differential Reovirus-Specific and Herpesvirus-Specific Activator Protein 1 Activation of Secretogranin II Leads to Altered Virus Secretion. Global medical discovery

 

 

Journal Reference

J Virol. 2015 Dec;89(23):11954-64.

Berard AR1, Severini A2, Coombs KM3.

Show Affiliations
  1. Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada Manitoba Center for Proteomics and Systems Biology, John Buhler Research Center, University of Manitoba, Winnipeg, MB, Canada.
  2. Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada.
  3. Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada Manitoba Center for Proteomics and Systems Biology, John Buhler Research Center, University of Manitoba, Winnipeg, MB, Canada Manitoba Institute of Child Health, University of Manitoba, Winnipeg, MB, Canada kevin.coombs@umanitoba.ca.

Abstract

Viruses utilize host cell machinery for propagation and manage to evade cellular host defense mechanisms in the process. Much remains unknown regarding how the host responds to viral infection. We recently performed global proteomic screens of mammalian reovirus TIL- and T3D-infected and herpesvirus (herpes simplex virus 1 [HSV-1])-infected HEK293 cells. The nonenveloped RNA reoviruses caused an upregulation, whereas the enveloped DNA HSV-1 caused a downregulation, of cellular secretogranin II (SCG2). SCG2, a member of the granin family that functions in hormonal peptide sorting into secretory vesicles, has not been linked to virus infections previously. We confirmed secretogranin II upregulation and found secretogranin II phosphorylation by 18 h postinfection (hpi) in reovirus-infected cells. We also found a decrease in the amount of reovirus secretion from secretogranin II knockdown cells. Similar analyses of cells infected with HSV-1 showed an increase in the amount of secreted virus. Analysis of the stress-activated protein kinase (SAPK)/Jun N-terminal protein kinase (JNK) pathway indicated that each virus activates different pathways leading to activator protein1 (AP-1) activation, which is the known SCG2 transcription activator. We conclude from these experiments that the negative correlation between SCG2 quantity and virus secretion for both viruses indicates a virus-specific role for secretogranin II during infection.

IMPORTANCE:

Mammalian reoviruses affect the gastrointestinal system or cause respiratory infections in humans. Recent work has shown that all mammalian reovirus strains (most specifically T3D) may be useful oncolytic agents. The ubiquitous herpes simplex viruses cause common sores in mucosal areas of their host and have coevolved with hosts over many years. Both of these virus species are prototypical representatives of their viral families, and investigation of these viruses can lead to further knowledge of how they and the other more pathogenic members of their respective families interact with the host. Here we show that secretogranin II (SCG2), a protein not previously studied in the context of virus infections, alters virus output in a virus-specific manner and that the quantity of SCG2 is inversely related to amounts of infectious-virus secretion. Herpesviruses may target this protein to facilitate enhanced virus release from the host.

Copyright © 2015, American Society for Microbiology. All Rights Reserved.

Go To J Virol

Comparative Tissue Proteomics of Microdissected Specimens Reveals Novel Candidate Biomarkers of Bladder Cancer

Significance Statement

This manuscript is focused on the biomarker discovery from clinical tissue specimens from bladder cancer patients as well as biomarker verification using several strategies. The results were integrated with a series of papers published by this group of urine proteomes [1-3] to select potential biomarker for further verification studies. Transgelin-2 (TAGLN2) and stathmin 1 (STMN1) proteins were successful verified with higher concentrations in bladder cancer tissue and urine specimens. The integrated results suggest that up-regulated TAGLN2 in bladder tumor cells may be secreted into the urine through microparticles, which in turn causes the higher concentration of urinary TAGLN2.

References:

  1. Chen, Y.T., et al., Discovery of novel bladder cancer biomarkers by comparative urine proteomics using iTRAQ technology. J. Proteome Res., 2010. 9(11): p. 5803-15.
  2. Chen, C.L., et al., Comparative and targeted proteomic analyses of urinary microparticles from bladder cancer and hernia patients. J. Proteome Res., 2012. 11(12): p. 5611-29.
  3. Chen, C.L., et al., Identification of potential bladder cancer markers in urine by abundant-protein depletion coupled with quantitative proteomics. J. Proteomics, 2013. 85C: p. 28-43.
 

About The Author

Dr. Yi-Ting Chen received her Ph.D. degree in Department of Chemistry from the National Tsing Hua University, Taiwan. Before graduation, she went to University of Alberta, Canada as a visiting student in 2001. Following the graduation in 2001, she worked as a research scientist in the Industrial Technology Research Institute, Taiwan to establish a new mass spectrometry lab for numerous proteomic projects. From September 2007, she worked at Molecular Medicine Research Center of Chang Gung University (CGU) as an associated research scientist, with focusing on urinary protein biomarker discovery in bladder cancer. Being an assistant professor in Department of Biomedical Sciences of CGU in 2013, Dr. Chen’s research interests focus on proteomics, metabolomics and systems biology of urological diseases and advanced method development for detection of biomolecules, primarily based on mass spectrometry.

 Comparative Tissue Proteomics of Microdissected Specimens Reveals Novel Candidate Biomarkers of Bladder Cancer. Global medical discovery

Journal Reference

Mol Cell Proteomics. 2015;14(9):2466-78.

Chien-Lun Chen1, Ting Chung2, Chih-Ching Wu3, Kwai-Fong Ng4, Jau-Song Yu 5, Cheng-Han Tsai 6, Yu-Sun Chang5, Ying Liang 2, Ke-Hung Tsui 1, Yi-Ting Chen7

Show Affiliations
  1. From the ‡Department of Urology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; §School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan;
  2. Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan;
  3. Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan; ‖Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan;
  4. Department of Pathology, Chang Gung Memorial Hospital, Taoyuan, Taiwan;
  5. Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan;
  6. Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan;
  7. Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan. ytchen@mail.cgu.edu.tw.


Abstract

More than 380,000 new cases of bladder cancer are diagnosed worldwide, accounting for ∼150,200 deaths each year. To discover potential biomarkers of bladder cancer, we employed a strategy combining laser microdissection, isobaric tags for relative and absolute quantitation labeling, and liquid chromatography-tandem MS (LC-MS/MS) analysis to profile proteomic changes in fresh-frozen bladder tumor specimens. Cellular proteins from four pairs of surgically resected primary bladder cancer tumor and adjacent nontumorous tissue were extracted for use in two batches of isobaric tags for relative and absolute quantitation experiments, which identified a total of 3220 proteins. A DAVID (database for annotation, visualization and integrated discovery) analysis of dysregulated proteins revealed that the three top-ranking biological processes were extracellular matrix organization, extracellular structure organization, and oxidation-reduction. Biological processes including response to organic substances, response to metal ions, and response to inorganic substances were highlighted by up-expressed proteins in bladder cancer. Seven differentially expressed proteins were selected as potential bladder cancer biomarkers for further verification. Immunohistochemical analyses showed significantly elevated levels of three proteins-SLC3A2, STMN1, and TAGLN2-in tumor cells compared with noncancerous bladder epithelial cells, and suggested that TAGLN2 could be a useful tumor tissue marker for diagnosis (AUC = 0.999) and evaluating lymph node metastasis in bladder cancer patients. ELISA results revealed significantly increased urinary levels of both STMN1 and TAGLN2 in bladder cancer subgroups compared with control groups. In comparisons with age-matched hernia urine specimens, urinary TAGLN2 in bladder cancer samples showed the largest fold change (7.13-fold), with an area-under-the-curve value of 0.70 (p < 0.001, n = 205). Overall, TAGLN2 showed the most significant overexpression in individual bladder cancer tissues and urine specimens, and thus represents a potential biomarker for noninvasive screening for bladder cancer. Our findings highlight the value of bladder tissue proteome in providing valuable information for future validation studies of potential biomarkers in urothelial carcinoma.

© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.

Go To Mol Cell Proteomics. 

Measles Virus Infection Inactivates Cellular Protein Phosphatase 5 with Consequent Suppression of Sp1 and c-Myc Activities

About The Author

Prof. Chieko Kai received her Ph.D in Veterinary Medical Science, The University of Tokyo in 1983. After that, she got a position of an Assistant Professor in Institute of Medical Science, The University of Tokyo, worked as a visiting fellow in Department of Tumor Biology, Karolinska Institute, Sweden between 1985-1987, and then obtained a position as an Associate Professor at Department of Veterinary Microbiology, Graduate School of Agriculture. In November 1999, she was appointed to the full professorship in Institute of Medical Science, The University of Tokyo.  She has also a professor position in the international Research Center for Infectious Diseases in Institute as well since 2005 till now, and also the Director of Laboratory Animal Research Center since Apr 2000. She was the Vice-Dean of the Institute from 2003 to 2004.  She is a member of Science Council of Japan. Her major research interests are to elucidate molecular mechanisms of pathogenicity and species specificity of negative and single strand RNA viruses (Mononegavirales), and to control viral diseases. For these purposes, her group is studying virus replication and identifying viral and host factors important for the expression of pathogenicity using a novel reverse genetics technique. Her group is also developing new virus vaccines to infectious diseases, and a novel cancer therapy using oncolytic viruses by genetic engineering.

About The Author

Dr. Hiroki Sato received his PhD from Chiba University in 1999, and joined Dr. Chieko Kai’s lab; Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo. He is an assistant professor and his current research focuses on the mechanisms of cell-type specific cellular responses and comprehensive network of post transcriptional modifications after morbillivirus infection.

Figure Legend

The intracellular accumulation of viral nucleocapsid causes the inactivation of PP5, autophosphorylation and the resulting inactivation of DNA-PKcs, then the downstream reduction of Sp1 phosphorylation and degradation of c-Myc, both of which cause their inactivation and consequent downregulation of housekeeping genes.

 

Measles Virus Infection Inactivates Cellular Protein Phosphatase 5 with Consequent Suppression of Sp1 and c-Myc Activities. Global medical discovery

Journal Reference

J Virol. 2015 Oct;89(19):9709-18.

Sato H1, Yoneda M1, Honma R2, Ikeda F1, Watanabe S2, Kai C3.

Show Affiliations
  1. Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
  2. Clinical Informatics, Tokyo Medical and Dental University, Tokyo, Japan.
  3. Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan ckai@ims.u-tokyo.ac.jp.

Abstract

Measles virus (MeV) causes several unique syndromes, including transient immunosuppression. To clarify the cellular responses to MeV infection, we previously analyzed a MeV-infected epithelial cell line and a lymphoid cell line by microarray and showed that the expression of numerous genes was up- or downregulated in the epithelial cells. In particular, there was a characteristic comprehensive downregulation of housekeeping genes during late stage infection. To identify the mechanism underlying this phenomenon, we examined the phosphorylation status of transcription factors and kinase/phosphatase activities in epithelial cells after infection. Measles virus infection inactivated cellular protein phosphatase 5 (PP5) that consequently inactivated DNA-dependent protein kinase, which reduced Sp1 phosphorylation levels, and c-Myc degradation, both of which downregulated the expression of many housekeeping genes. In addition, intracellular accumulation of viral nucleocapsid inactivated PP5 and subsequent downstream responses. These findings demonstrate a novel strategy of Measles virus during infection, which causes the collapse of host cellular functions.

IMPORTANCE:

Measles virus (MeV) is one of the most important pathogens in humans. We previously showed that Measles virus infection induces the comprehensive downregulation of housekeeping genes in epithelial cells. By examining this phenomenon, we clarified the molecular mechanism underlying the constitutive expression of housekeeping genes in cells, which is maintained by cellular protein phosphatase 5 (PP5) and DNA-dependent protein kinase. We also demonstrated that MeV targets PP5 for downregulation in epithelial cells. This is the first report to show how MeV infection triggers a reduction in overall cellular functions of infected host cells. Our findings will help uncover unique pathogenicities caused by Measles virus.

Copyright © 2015, American Society for Microbiology. All Rights Reserved.

Go To J Virol

 

Thursday, February 18, 2016

Methodologic quality assessment of red blood cell transfusion guidelines and the evidence base of more restrictive transfusion thresholds

Significance Statement

The World Health Organization indicates that every second someone in the world needs a blood transfusion. Blood transfusions therefore save many lives every year, and patients can die when they don’t receive a transfusion when necessary. However, transfusions also carry certain risks, and therefore should not be performed unless really necessary.

In recent years, a lot of emphasis has been put on this second consideration, which has been more broadly rebranded as “patient blood management” (PBM). PBM is a worthwhile effort, but should be evidence-based and not based on other considerations, be it religious (e.g., refusing of an allogenic blood transfusion as a Jehovah Witness) or commercial (e.g., receiving sponsorships by commercial companies who promote RBC replacing products).

It is therefore important that transfusion guidelines are of high quality, and based on the best scientific evidence. Our analysis of the quality of the most widely used transfusion guidelines, using an internationally used tool (AGREE II), indicates that guidelines recommending red blood cell transfusion as of Hb levels of 7 to 8 g/dL are based on high quality evidence, whereas the more recent guidelines recommending red blood cell transfusion only at Hb thresholds of ≤6 g/dL are not based on solid evidence.

The methodological robustness of the published transfusion guidelines is variable and has room for improvement. Furthermore, guideline developers and reviewers should always be transparent about conflicts of interest, whether religious or commercial, which was, based on other available information, not always the case in these guidelines.

About The Author

Hans Van Remoortel, PT/PhD, physiotherapist, is staff member in the Centre for Evidence-Based Practice of the Belgian Red Cross-Flanders. After obtaining a Master in Rehabilitation Sciences and Physiotherapy in 2005, Hans worked for three years (2005-2008) as a clinical physiotherapist in cardiac rehabilitation programs in 2 Belgian hospitals (University Hospital of Leuven and Imelda Hospital Bonheiden). After working in a clinical setting, he obtained his PhD degree at the Faculty of Rehabilitation Sciences and Kinesiology at the University of Leuven (Belgium) with a project entitled ‘Physical activity and comorbidities in patients with chronic obstructive pulmonary disease’ (2009-2013).

He has published about 20 articles in peer reviewed journals, mainly in the field of exercise, physical activity, pulmonology and evidence-based medicine. In his current job, Hans is a methodologist working on the development of systematic reviews and guidelines within the relevant themes of the Belgian Red Cross, i.e. from humanitarian aid to blood supply. In his spare moments, Hans enjoys to spend time with his family, is a recreational sporter (squash, soccer, running, cycling) and enjoys to attend live music concerts.

About The Author

Philippe Vandekerckhove, M.D./PhD, pathologist, is the CEO of the Belgian Red Cross-Flanders.

Prior to this position Philippe worked as Clinical Director of the University Hospital Leuven from where he also obtained his M.D./PhD and Pathology degree. His clinical and pathology training was further carried out in South Africa (Baragwanath – University of Johannesburg, and Groote Schuur Hospital – University of Cape Town), the US (Woods Hole Marine Biology Laboratory, University of Hawaii, New York University), and The Netherlands (Erasmus University, Rotterdam).

In addition, Philippe studied healthcare management at INSEAD (France) and general management at Harvard Business School. He has published about 60 articles in peer reviewed journals, and 5 chapters in textbooks, mainly in the field of immunology, hematology, blood banking and evidence-based medicine.

Philippe is associate professor at the Faculties of Medicine of the University of Leuven and the University of Ghen. He holds non-executive positions as president of the European Blood Alliance, president of GAP, member of the governing board of the International Federation of Red Cross and Red Crescent Societies, and of the investment committee of Flanders’ Care Invest (Flemish government). In his spare moments, he enjoys jogging, natural horsemanship and time and travel spent in natural surroundings.

Methodologic quality assessment of red blood cell transfusion guidelines and the evidence base of morerestrictive transfusion thresholds.

Journal Reference

Transfusion. 2016 Feb;56(2):472-80.

Van Remoortel H1, De Buck E1, Dieltjens T1, Pauwels NS1, Compernolle V1,2, Vandekerckhove P1,2,3.

Show Affiliations
  1. BelgianRed Cross-Flanders, Mechelen, Belgium.
  2. Faculty of Medicine, University of Ghent, Ghent, Belgium.
  3. Department of Public Health and Primary Care, Faculty of Medicine, Catholic University of Leuven, Leuven, Belgium.

Abstract

BACKGROUND:

Recent literature suggests that more restrictive red blood cell (RBC) transfusion practices are equivalent or better than more liberal transfusion practices. The methodologic quality of guidelines recommending more restrictive transfusion thresholds and their underlying scientific evidence is unclear. Therefore, we aimed to evaluate the quality of the development process of red blood cell transfusion guidelines and to investigate the underlying evidence of guidelines recommending a more restrictive hemoglobin (Hb) threshold.

STUDY DESIGN AND METHODS:

Via systematic literature screening of relevant databases (NGC, GIN, Medline, and Embase), red blood cell transfusion guidelines recommending a more restrictive Hb level (<6, <7, or <8 g/dL) were included. Four assessors independently evaluated the methodologic quality by scoring the rigor of development domain (AGREE II checklist). The level of evidence served as a reference for the quality of the underlying evidence.

RESULTS:

The methodologic quality of 13 red blood cell transfusion guidelines was variable (18%-72%) but highest for those developed by Advancing Transfusion and Cellular Therapies Worldwide (72%), the Task Force of Advanced Bleeding Care in Trauma (70%), and the Dutch Institute for Healthcare Improvement (61%). A Hb level of less than 7 g/dL (intensive care unit patients) or less than 8 g/dL (postoperative patients) were the only thresholds based on high-quality evidence. Only four of 32 recommendations had a high-quality evidence base.

CONCLUSION:

Methodologic quality should be guaranteed in future RBC transfusion guideline development to ensure that the best available evidence is captured when recommending restrictive transfusion strategies. More high-quality trials are needed to provide a stronger scientific basis for RBC transfusion guidelines that recommend more restrictive transfusion thresholds.

© 2015 AABB.

Go To Transfusion

 

 

Modulation of the gene expression of annulus fibrosus-derived stem cells using poly(ether carbonateurethane)urea scaffolds of tunable elasticity

Significance Statement

Annulus fibrosus (AF) injuries lead to substantial intervertebral disc deterioration which characterizes degenerative disc diseases. Repair of AF, however, remains challenging due to the tremendous heterogeneity of Annulus fibrosus tissue. Since the differentiation of stem cells significantly relies on the elasticity of substrate, a series of biodegradable poly(ether carbonate urethane)urea (PECUU) materials whose elasticity resembled that of native Annulus fibrosus tissue were synthesized in this study. When Annulus fibrosus-derived stem cells (AFSCs) were cultured on electrospun PECUU fibrous scaffolds, the gene expression and protein production of major matrix components, including collagen-I, collagen-II and aggrecan, and cell traction forces gradually changed with the elasticity of PECUU. Such substrate elasticity-dependent modulation of Annulus fibrosus-derived stem cells was similar to the gradual transition in the genetic, biochemical, and biomechanical characteristics of cells from inner to outer regions of native Annulus fibrosus tissue. This work has, for the first time, revealed that Annulus fibrosus-derived stem cells are able to present different gene expression patterns simply as a result of the elasticity of scaffold material. Findings from this study will help develop adequate materials for Annulus fibrosus regeneration.

About The Author

Professor Bin Li is the director of the Biomaterials and Cell Mechanics Laboratory (BCML) of Orthopedic Institute at Soochow University, Suzhou, China. He received the bachelor degree in 1996 and PhD degree in Materials Science from Tsinghua University in 2001. He then joined the Institute of Materials Research and Engineering, Singapore as a Research Associate until 2004. After that he consecutively pursued research training at Carnegie Mellon University, University of Pittsburgh, and Harvard University until 2009, when he took the current position as a full professor at Soochow University. He is the recipient of the Orthopaedics Research Award (1st prize) from Chinese Orthopaedic Association, Xu Guangqi Program from the French Embassy in China, and France Talent Innovation from the Consulate General of France in Shanghai. He currently serves as the chair of China Development Committee of International Chinese Musculoskeletal Research Society (ICMRS). He is a fellow of Chinese Orthopaedic Research Society (CORS), Chinese Association of Orthopaedic Surgeons (CAOS), Chinese Association of Rehabilitation Medicine (CARM), and International Society of Orthopaedic Surgery and Traumatology (SICOT). He has delivered about 40 invited and is the author of over 70 publications and 9 book chapters. He now leads a multidisciplinary research group studying biomaterials for bone and cartilage repair, stem cells and tissue engineering, smart molecular recognition and controlled release, surface modification and functionalization, and cellular biomechanics and mechanobiology.

About The Author

Dr. Caihong Zhu is a research associate and a member of the Biomaterials and Cell Mechanics Laboratory (BCML) of Orthopedic Institute at Soochow University. She received the bachelor degree in Polymer Chemistry from Nanjing University in 2001 and PhD degree in Polymer Chemistry from Soochow University in 2010. She joined Soochow University in 2012. Her research interests include biomaterials for bone and intervertebral disc regeneration and controlled drug delivery.

  

About The Author

Dr. Jun Li is an orthopaedic surgeon who received his PhD degree at the Soochow University. Currently, he is a postdoctoral researcher at Shenzhen University. His major research interests include bone and cartilage regeneration via stem cell transplantation and osteoporosis.

Modulation of gene expression of annulus fibrosus-derived stem cells using poly(ether carbonateurethane)urea scaffolds of tunable elasticity. Global Medical Discovery

Journal Reference

Acta Biomater. 2016;29:228-38.

Caihong Zhu, Jun Li, Chen Liu, Pinghui Zhou, Huilin Yang, Bin Li.

Department of Orthopaedics, The First Affiliated Hospital, Orthopaedic Institute, Soochow University, 188 Shizi St, Suzhou, Jiangsu 215006, China.

These authors contributed equally to this study. Email: binli@suda.edu.cn

Abstract

Annulus fibrosus (AF) injuries commonly lead to substantial deterioration of the intervertebral disc (IVD). While tissue engineering has recently evolved into a promising approach for AF regeneration, it remains challenging due to the cellular, biochemical, and mechanical heterogeneity of AF tissue. In this study, we explored the use of AF-derived stem cells (AFSCs) to achieve diversified differentiation of cells for AF tissue engineering. Since the differentiation of stem cells relies significantly on the elasticity of the substrate, we synthesized a series of biodegradable poly(ether carbonate urethane)urea (PECUU) materials whose elasticity approximated that of native AF tissue. When AFSCs were cultured on electrospun PECUU fibrous scaffolds, the gene expression of collagen-I in the cells increased with the elasticity of scaffold material, whereas the expression of collagen-II and aggrecan genes showed an opposite trend. At the protein level, the content of collagen-I gradually increased with substrate elasticity, while collagen-II and GAG contents decreased. In addition, the cell traction forces (CTFs) of AFSCs gradually decreased with scaffold elasticity. Such substrate elasticity-dependent changes of AFSCs were similar to the gradual transition in the genetic, biochemical, and biomechanical characteristics of cells from inner to outer regions of native AF tissue. Together, findings from this study indicate that AFSCs, depending on the substrate elasticity, have strong tendencies to differentiate into various types of AF-like cells, thereby providing a solid foundation for the tissue engineering applications of AFSCs.

Copyright © 2016. Published by Elsevier B.V.

Go To Acta Biomater 

 

 

Human telomerase reverse transcriptase binds to a pre-organized hTR in vivo exposing its template

About The Author

Dr. Georgeta Zemora studied Biochemistry at the Gene Center, Ludwig Maximilians University, Munich. She completed her PhD in Molecular Biology at the University of Vienna. During her PhD at MFPL Vienna she was guided by her mentor, Dr Christina Waldsich who introduced her into the fascinating field of RNA folding in vivo. Her research focused on deciphering the structure of human telomerase RNA (hTR) in vivo as well as its interaction with the human telomerase reverse transcriptase (hTERT). 

Journal Reference

Nucleic Acids Res. 2016 Jan 8;44(1):413-25.

Zemora G1, Handl S2, Waldsich C2.

Show Affiliations
  1. Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Dr Bohrgasse 9/5, A-1030 Vienna, Austria georgeta.zemora@univie.ac.at.
  2. Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories, University of Vienna, Dr Bohrgasse 9/5, A-1030 Vienna, Austria.

Abstract

Telomerase is a specialized reverse transcriptase that is responsible for telomere length maintenance. As in other organisms, the minimal components required for an active human telomerase are the template-providing telomerase RNA (hTR) and the enzymatic entity telomerase reverse transcriptase (hTERT). Here, we explored the structure of hTR and the hTERT-induced conformational changes within hTR in living cells. By employing an in vivo DMS chemical probing technique, we showed that the pseudoknot and associated triple helical scaffold form stably in vivo independently of hTERT. In fact, the dimethyl-sulfate (DMS) modification pattern suggests that hTR alone is capable of adopting a conformation that is suited to interact with hTERT. However, in the absence of hTERT the template region of hTR is only weakly accessible to DMS-modifications. The predominant change after binding of hTERT to hTR is the exposure of the template region.

© The Author(s) 2015. Published by Oxford University Press on behalf of Nucleic Acids Research.

Go To Nucleic Acids Res

Suppression of Th2 and Tfh immune reactions by Nr4a receptors in mature T reg cells

Significance Statement

Immune systems protect our body from threats by various infectious pathogens, including bacteria, virus, helminth, and fungi. In the immune systems, CD4+ helper T cells (Th) play central roles to exert appropriate immune reactions to such innumerable sorts of pathogens. However, if the function of Th are wrongly directed to self-antigens or non-harmful antigens, including pollens, food antigens, and commensals, it will be concluded to serious inflammatory diseases like autoimmune diseases and allergies. On the other hand, immune system has an important cell subset, regulatory T (Treg) cells, which suppress such wrong immune reactions. Treg cells have been known to be critical in suppression of inflammatory diseases, however, how Treg cell lineage was properly maintained in our body had not been clarified in depth yet. In this study, we discovered that Nr4a family of nuclear orphan receptors play critical roles in the maintenance of the Treg cell lineage. Nr4a-deficient Treg cells showed a global reduction of Treg cell-associated gene expressions, compared with those in wildtype Treg cells. Although wildtype Treg cells properly suppressed immune reactions elicited by Th2 and Tfh cells, it was revealed that Nr4a-deficient Treg cells have lost such suppressive activities. We also found that Nr4a-deficient Treg cells aberrantly convert to cells which have characteristics of inflammatory Th cells. Mice in which Nr4a factors were deleted specifically in Treg cells develop symptoms including allergic asthma and autoimmune diseases. Collectively, our work revealed that Nr4a factors play essential roles in Treg cell biology, thus present attractive therapeutic targets for treatment of various immune disorders, including atopic allergies and asthma. 

About The Author

Dr. Takashi Sekiya received his Bachelor of Science degree in 1999 from University of Tokyo. He obtained his Ph.D. from Graduate School of Agricultural and Life Sciences, The University of Tokyo in 2004. Then, he moved to Fox Chase Cancer Center as a postdoctoral fellow at Dr. Ken Zaret’s laboratory. In 2008, he moved back to Japan, and joined Dr. Akihiko Yoshimura’s lab in Keio University School of Medicine, there he has been working on mechanisms of immune tolerance, focusing on developmental programs of CD4 T cells. 

Suppression of Th2 and Tfh immune reactions by Nr4a receptors in mature T reg cells. Global Medical Discovery

Journal Reference

J Exp Med. 2015;212(10):1623-40.

Sekiya T1, Kondo T2, Shichita T2, Morita R2, Ichinose H3, Yoshimura A4.

Show Affiliations
  1. Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan yoshimura@a6.keio.jpt-sekiya@z7.keio.jp.
  2. Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan.
  3. Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8501, Japan.
  4. Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan Japan Science and Technology Agency (JST), Core Research for Evolutional Science and Technology, Chiyoda-ku, Tokyo 102-0075, Japan yoshimura@a6.keio.jpt-sekiya@z7.keio.jp.

Abstract

Regulatory T (T reg) cells are central mediators of immune suppression. As such, T reg cells are characterized by a distinct pattern of gene expression, which includes up-regulation of immunosuppressive genes and silencing of inflammatory cytokine genes. Although an increasing number of transcription factors that regulate T reg cells have been identified, the mechanisms by which the T reg cell-specific transcriptional program is maintained and executed remain largely unknown. The Nr4a family of nuclear orphan receptors, which we recently identified as essential for the development of T reg cells, is highly expressed in mature T reg cells as well, suggesting that Nr4a factors play important roles even beyond T reg cell development. Here, we showed that deletion of Nr4a genes specifically in T reg cells caused fatal systemic immunopathology. Nr4a-deficient Treg cells exhibited global alteration of the expression of genes which specify the T reg cell lineage, including reduction of Foxp3 and Ikzf4. Furthermore, Nr4a deficiency abrogated T reg cell suppressive activities and accelerated conversion to cells with Th2 and follicular helper T (Tfh) effector-like characteristics, with heightened expression of Th2 and Tfh cytokine genes. These findings demonstrate that Nr4a factors play crucial roles in mature T reg cells by directly controlling a genetic program indispensable for T reg cell maintenance and function.

© 2015 Sekiya et al.

Go To J Exp Med

 

 

The development of statin-based therapy for patients with hepatitis C virus (HCV) infection using human induced pluripotent stem (iPS) cell technology

Significance Statement

Human induced pluripotent stem cells (hiPSCs) have been expected to transform drug discovery by providing physiologically relevant cells for toxic compound identification and compound screening. When Shimada M et al. (J Hepatol. 2012;56: 299-300) considered two reports (Moriguchi H et al.; Hepatology. 2010; 51: 344–345 and 351–352) published in 2010 investigating the antiviral efficacy of pitavastatin against hepatitis C virus (HCV) infection in vitro, they conducted a randomized controlled trial (J Hepatol. 2012;56: 299-300). As a result, the results of 2010 proof-of-concept studies of the antiviral efficacies and safety of pitavastatin against HCV infection using a replicon system and human hepatocyte-like cells from hiPSCs (Moriguchi H et al.; Hepatology. 2010; 51: 344–345 and 351–352) were confirmed in a randomized controlled trial published by Shimada M et al. in 2012 (J Hepatol. 2012;56: 299-300). Therefore, this series of studies (Moriguchi H et al.; Hepatology. 2010; 51: 344–345 and 351–352, Shimada M et al.; J Hepatol. 2012; 56: 299-300) would be the first to report clinical applications of hiPSCs. Furthermore, after the clinical trial by Shimada M et al. (J Hepatol. 2012; 56: 299-300), the antiviral efficacies and safeties of pitavastatin against HCV infection were more confirmed in the two clinical studies (Kohjima M et al.; J Med Virol. 2013: 85: 250-260 and Yokoyama S et al.; Aliment Pharmacol Ther. 2014; 39: 443-444). Therefore, to investigate the antiviral efficacies and safety of pitavastatin against HCV infection using a replicon system and human hepatocyte-like cells from hiPSCs (Moriguchi H et al.; Hepatology. 2010; 51: 344–345 and 351–352) would be a rational approach as new drug discovery. 

About The Author

Dr. Moriguchi H obtained his Ph.D. from the University of Tokyo in 2007. He was a visiting associate professor (full time) from 2000 to 2006 and a project professor from 2006 to 2009 at the University of Tokyo. He is currently at Oak Clinic company. His research focuses on clinical epidemiology (1996, Gut, etc), clinical decision analysis (2012, Hepatology, etc), stem cell research (2011, Cell Mol Life Sci, etc) and reproductive medicine (2012, Hum Reprod, etc) in Gastroenterology, Hepatology, cancers, stem cell research and reproductive medicine.

Journal Reference

Clin Res Hepatol Gastroenterol. 2015;39(5):541-3.

Moriguchi H.

Oak Clinic, 2-7-9, Tamade-Nishi, Nishinari-ku, 557-0045 Osaka, Japan. Electronic address: moriguchi_h@oakclinic-group.com.

Abstract

Human induced pluripotent stem (iPS) cells may transform drug discovery. Here I show an example of the development of statin (HMG-CoA reductase inhibitors)-based therapy for patients with hepatitis C virus (HCV) infection using human iPS cell technology. When Shimada et al. considered the two reports on the antiviral effects of pitavastatin for HCV infection in vitro by Moriguchi et al., they conducted a randomized controlled trial. As a result, a proof-of-concept for the antiviral effect of pitavastatin against HCV infection using human iPS cell technology by Moriguchi et al. was confirmed in the randomized controlled trial by Shimada et al. in 2012. Therefore, above-mentioned a series of studies became to the first to report the clinical application of human iPS cells. Furthermore, here I propose that new clinical research methods using human iPS cell technology will be able to circumvent the limitations of conventional randomized controlled trials (RCTs) for the purpose of personalized medicine in the clinical setting.

Copyright © 2015 Elsevier Masson SAS. All rights reserved.

Go To Clin Res Hepatol Gastroenterol

Oxidative Stress and Response to Thymidylate Synthase-Targeted Antimetabolites

Journal Reference

Mol Pharmacol. 2015 Dec;88(6):970-81.

Ozer U1, Barbour KW1, Clinton SA1, Berger FG2.

Show Affiliations
  1. Department of Biological Sciences, and Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina.
  2. Department of Biological Sciences, and Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina fgberger@mailbox.sc.edu.

Abstract

Thymidylate synthase (TYMS; EC 2.1.1.15) catalyzes the reductive methylation of 2′-deoxyuridine-5′-monophosphate (dUMP) by N(5),N(10)-methyhlenetetrahydrofolate, forming dTMP for the maintenance of DNA replication and repair. Inhibitors of Thymidylate synthase have been widely used in the treatment of neoplastic disease. A number of fluoropyrimidine and folate analogs have been developed that lead to inhibition of the enzyme, resulting in dTMP deficiency and cell death. In the current study, we have examined the role of oxidative stress in response to TYMS inhibitors. We observed that intracellular reactive oxygen species (ROS) concentrations are induced by these inhibitors and promote apoptosis. Activation of the enzyme NADPH oxidase (NOX), which catalyzes one-electron reduction of O2 to generate superoxide (O2 (●-)), is a significant source of increased ROS levels in drug-treated cells. However, gene expression profiling revealed a number of other redox-related genes that may contribute to ROS generation. Thymidylate synthase inhibitors also induce a protective response, including activation of the transcription factor nuclear factor E2-related factor 2 (NRF2), a critical mediator of defense against oxidative and electrophilic stress. Our results show that exposure to Thymidylate synthase inhibitors induces oxidative stress that leads to cell death, while simultaneously generating a protective response that may underlie resistance against such death.

Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics.

Go To Mol Pharmacol

 

 

Effective use of the TSPY gene-specific copy number in determining fetal DNA in the maternal blood of cynomolgus monkeys

Journal Reference

Anim Sci J. 2015 Sep 30.

Yasmin L, Takano JI, Sankai T.

Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Japan.

Abstract

Since the available concentration of single-copy fetal genes in maternal blood DNA is sometimes lower than detection limits by PCR methods, the development of specific and quantitative PCR detection methods for fetal DNA in maternal blood is anticipated, which may broaden the methods that can be used to monitor pregnancy. We used the TaqMan qPCR amplification for DYS14 multi-copy sequence and the SRY gene in maternal blood plasma (cell-free DNA) and fractional precipitated blood cells (cellular DNA) from individual cynomolgus monkeys at 22 weeks of pregnancy. The availability of cell-free fetal DNA was higher in maternal blood plasma than that of cellular DNA from fractional precipitated blood cells. There was a significantly higher (P < 0.001) mean copy number of fetal male DYS14 from maternal plasma (4.4 × 104 copies/mL) than that of detected fetal cellular DNA from fractional blood cell pellets. The sensitivity of the DYS14 PCR assay was found to be higher than that of the SRY assay for the detection of fetal DNA when its presence was at a minimum. The DYS14 assay is an improved method for quantifying male fetal DNA in circulating maternal blood in the primate model.

© 2015 Japanese Society of Animal Science.

Go To Anim Sci J

 

 

 

Effect of decellularized tissue powders on a rat model of acute myocardial infarction

Journal Reference

Mater Sci Eng C Mater Biol Appl. 2015 Nov 1;56:494-500.

Tabuchi M1, Negishi J2, Yamashita A3, Higami T1, Kishida A4, Funamoto S5. 

Show Affiliations
  1. Department of Thoracic and Cardiovascular Surgery, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo 060-8543, Japan.
  2. Department of Thoracic and Cardiovascular Surgery, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo 060-8543, Japan; Japan Society for the Promotion of Science 8 Ichibancho, Chiyoda-ku, Tokyo 102-8472, Japan; Department of Material-based Medical Engineering, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10, Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
  3. Department of Thoracic and Cardiovascular Surgery, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo 060-8543, Japan; Department of Material-based Medical Engineering, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10, Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
  4. Department of Material-based Medical Engineering, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10, Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
  5. Department of Thoracic and Cardiovascular Surgery, Sapporo Medical University School of Medicine, South of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10, Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan. Electronic address: sfunamoto.atrm@tmd.ac.jp.

Abstract

Many research groups are currently investigating new treatment modalities for myocardial infarction. Numerous aspects need to be considered for the clinical application of these therapies, such as low cell integration and engraftment rates of cell injection techniques. Decellularized tissues are considered good materials for promoting regeneration of traumatic tissues. The properties of the decellularized tissues are sustained after processing to powder form. In this study, we examined the use of decellularized tissue powder in a rat  model of acute myocardial infarction.  The decellularized tissue powders, especially liver powder, promoted cell integration and neovascularization both in vitro and in vivo. Decellularized liver powder induced neovascularization in the infarct area, resulting in the suppression of myocardial necrosis. The results of this study suggest that decellularized liver powder has good potential for application as a blood supply material for the treatment of myocardial infarction.

Copyright © 2015 Elsevier B.V. All rights reserved.

Go To Mater Sci Eng C Mater Biol Appl

 

Roles of the canonical myomiRs miR-1, -133 and -206 in cell development and disease

Significance Statement

A large body of research has shown the importance of the canonical myomiRs in muscle development and maintenance, but more recently mis-expression of one or more myomiRs has been found important in a variety of cancers.  Whilst most myomiR dysregulation in cancer suggests a tumor-suppressor function, in a significant number of cancers a tumor-enhancer role is seen. This duality of roles is similar to that seen with a number of other important cancer-related miRNAs in different cancer types. This review collates the large body of evidence of myomiR functions in a wide variety of normal tissues and organs and relates these functions to their biological effects on validated target genes during the enhancement of cancer progression.

About The Author

KEITH MITCHELSON Has undertaken research in molecular biology, genetics, and biotechnology in the UK, Australia and China.  Until recently undertook research and business at the National Engineering Research Centre for Biochip Technology in Beijing. 

About The Author

WENYAN QIN Has undertaken research in biogenesis and progression of cancer at Tsinghua University School of Medicine, China. Currently undertake research at Capitalbio Technology Co. Ltd. 

Journal Reference

World J Biol Chem. 2015;6(3):162-208.

Mitchelson KR, Qin WY.

Keith Richard Mitchelson, Wen-Yan Qin, National Engineering Research Centre for Beijing Biochip Technology, Beijing 102206, China.

Abstract

MicroRNAs are small non-coding RNAs that participate in different biological processes, providing subtle combinational regulation of cellular pathways, often by regulating components of signalling pathways. Aberrant expression of miRNAs is an important factor in the development and progression of disease. The canonical myomiRs (miR-1, -133 and -206) are central to the development and health of mammalian skeletal and cardiac muscles, but new findings show they have regulatory roles in the development of other mammalian non-muscle tissues, including nerve, brain structures, adipose and some specialised immunological cells. Moreover, the deregulation of myomiR expression is associated with a variety of different cancers, where typically they have tumor suppressor functions, although examples of an oncogenic role illustrate their diverse function in different cell environments. This review examines the involvement of the related myomiRs at the crossroads between cell development/tissue regeneration/tissue inflammation responses, and cancer development.

Go To World J Biol Chem

Modeling and Prediction of Solvent Effect on Human Skin Permeability using Support Vector Regression and Random Forest

Journal Reference

Pharm Res. 2015 Nov;32(11):3604-17.

Baba H1,2, Takahara J3, Yamashita F4, Hashida M4,5

Show Affiliations
  1. Kyoto R&D Center, Maruho Co., Ltd., 93 Awata-cho, Chudoji, Shimogyo-ku, 600-8815, Kyoto, Japan. baba_dfq@mii.maruho.co.jp.
  2. Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29, Yoshida-shimoadachicho, Sakyo-ku, Kyoto, 606-8501, Japan. baba_dfq@mii.maruho.co.jp.
  3. Kyoto R&D Center, Maruho Co., Ltd., 93 Awata-cho, Chudoji, Shimogyo-ku, 600-8815, Kyoto, Japan.
  4. Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29, Yoshida-shimoadachicho, Sakyo-ku, Kyoto, 606-8501, Japan.
  5. Institute for Integrated Cell-Material Sciences, Kyoto University, 46-29, Yoshida-shimoadachicho, Sakyo-ku, Kyoto, 606-8501, Japan.

Abstract

PURPOSE:

The solvent effect on skin permeability is important for assessing the effectiveness and toxicological risk of new dermatological formulations in pharmaceuticals and cosmetics development. The solvent effect occurs by diverse mechanisms, which could be elucidated by efficient and reliable prediction models. However, such prediction models have been hampered by the small variety of permeants and mixture components archived in databases and by low predictive performance. Here, we propose a solution to both problems.

METHODS:

We first compiled a novel large database of 412 samples from 261 structurally diverse permeants and 31 solvents reported in the literature. The data were carefully screened to ensure their collection under consistent experimental conditions. To construct a high-performance predictive model, we then applied   support vector regression (SVR) and random forest (RF) with greedy stepwise descriptor selection to our database. The models were internally and externally validated.

RESULTS:

The support vector regression achieved higher performance statistics than random forest. The (externally validated) determination coefficient, root mean square error, and mean absolute error of support vector regression were 0.899, 0.351, and 0.268, respectively. Moreover, because all descriptors are fully computational, our method can predict as-yet unsynthesized compounds.

CONCLUSION:

Our high-performance prediction model offers an attractive alternative to permeability experiments for pharmaceutical and cosmetic candidate screening and optimizing skin-permeable topical formulations.

Go To Pharm Res

Friday, February 5, 2016

Long-term safety and activity of cladribine in patients with extranodal B-cell marginal zone lymphoma of the mucosa-associated lymphoid tissue (MALT) lymphoma

About The Author

Prof. Markus Raderer, M.D.

Medical University of Vienna, Vienna, Austria

Markus Raderer undertook his early medical studies at the Medical University of Vienna, completing his residency in internal medicine between 1993 and 1999, specialising in haematology and oncology in 2001. He received the venia docendi for ‘Applied and Experimental Oncology’ and ‘Internal Medicine’ in 1999 and 2002, respectively.  Professor Raderer is currently the Programme Director for Extranodal Lymphomas and for Endocrine Tumours at the Medical University of Vienna. He has been responsible for the management and conduct of phase II and III clinical trials at his institution since 1992.

Professor Raderer is on the Editorial Board of a number of journals including the Journal of Clinical Oncology, World Journal of Gastroenterology, and Middle European Journal of Medical Oncology. He has participated on consensus panels for the European Gastrointestinal Lymphoma Study Group (EGILS) on the topic of B-cell lymphoma of mucosa-associated lymphoid tissue, and the European Society of Medical Oncology (ESMO) on the subject of the management of lymphoid malignancies.

For a list of publications see http://ift.tt/1R9XUti

About The Author

Barbara Kiesewetter, M.D.

Medical University of Vienna, Vienna, Austria

Barbara Kiesewetter undertook her early medical studies at the Medical University of Vienna. Currently she is doing her residency in internal medicine at the Clinical Division of Oncology, Department of Medicine I, Medical University of Vienna, and is a PhD student of the local doctoral program of “Applied Clinical Science”. Dr. Kiesewetter is part of the study group of Prof. Raderer (Extranodal Lymphomas and Endocrine Tumours) since 2010 and was involved in recent clinical trials and scientific work.

For a list of publications see http://ift.tt/1R9XUJA  

Journal Reference

Hematol Oncol. 2015 Nov 18.

Kiesewetter B1, Dolak W2, Simonitsch-Klupp I3, Mayerhoefer ME4, Raderer M1.

Show Affiliations
  1. Department of Internal Medicine I, Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria.
  2. Department of Internal Medicine III, Clinical Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria.
  3. Department of Pathology, Medical University of Vienna, Vienna, Austria.
  4. Department of Radiology, Medical University of Vienna, Vienna, Austria.

Abstract

The purine analogue 2-chloro-deoxyadenosine (2-CDA, cladribine) +/- rituximab has been successfully tested in mucosa-associated lymphoid tissue lymphoma (MALT lymphoma) patients. However, studies using cladribine in other indications have reported the potential for prolonged hematological side effects and secondary hematologic and non-hematologic malignancies. To date, there have been no data on long-term effects of cladribine inMALT  lymphoma patients. We have analyzed a large number of 49 patients treated with cladribine at our institution 1997-2011. All patients were treated within clinical trials and had undergone a standardized follow-up protocol minimizing a potential bias in the detection of late sequels and relapses. After a median follow-up time of 61 months (interquartile range: 43-72) for 49 analyzed patients, 35 (71%) are alive, while 14 (29%) have died. In the entire collective, three cases (6%) of prolonged pancytopenia including manifest myelodysplastic syndrome in one patient (2%), three cases (6%) of secondary lymphoid malignancies, and five cases (10%) of non-hematologic cancers were documented. In terms of outcome, 42/49 (86%) patients responded to cladribine-containing treatment, and only 10/42 (24%) responding patients needed further treatment after a median time to progression of 14 months (interquartile range, 8-34). Currently, 25/35 (71%) patients being alive are in ongoing complete remission and 2/35 (6%) in ongoing stable disease, respectively. Eight patients (23%) are free of lymphoma after second-line therapy, with the median overall survival not having been reached. Our data suggest that cladribine might be safely applied in patients with MALT lymphoma, also in terms of long-term toxicities. These data also confirm the potential of cladribine to induce durable remissions.

Copyright © 2015 John Wiley & Sons, Ltd.

Go To Hematol Oncol

Significance information is found in the file below: 

Long-term safety and activity of cladribine in patients with extranodal B-cell marginal zone lymphoma of the mucosa-associated lymphoid tissue (MALT) lymphoma