Investigating the Molecular Mechanisms of Action of Lenalidomide and Other Immunomodulatory Derivatives

Total Page:16

File Type:pdf, Size:1020Kb

Investigating the Molecular Mechanisms of Action of Lenalidomide and Other Immunomodulatory Derivatives Investigating the Molecular Mechanisms of Action of Lenalidomide and Other Immunomodulatory Derivatives The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Haldar, Saurav Daniel. 2018. Investigating the Molecular Mechanisms of Action of Lenalidomide and Other Immunomodulatory Derivatives. Doctoral dissertation, Harvard Medical School. Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:37006480 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA TABLE OF CONTENTS! ! 1 Abstract……………………………………………………………………………………………..3 2 Glossary……………………………………………………………………………………………..5 3 Introduction………………………………………………………………………………………....6 3.1 A Tragic Beginning…...………………………………………………………………………..6 3.2 Thalidomide: The Comeback Drug…………………………………………………………..9 3.3 Development of Thalidomide Analogs……………………………………………………..12 3.4 iMiD Mechanism of Action…………………………………………………………………..15 3.5 Purpose of Inquiry……………………………………………………………………………22 4 Materials and Methods…………………………………………………...…...…………………24 4.1 Mutant Degron Analysis via a Fluorescent Reporter Assay……….……….…….…......24 4.2 Generation of CRISPR/Cas9 Single Cell Knockout Clones………………….…….……25 4.3 Western Immunoblotting…………………………………………………………………….28 4.4 TNF-α Enzyme-Linked Immunosorbent Assay (ELISA)……………………..………….29 5 Results.……………………………………………………………………………….……………31 5.1 Effects of IKZF3 Degron Mutagenesis on Substrate Recognition by the iMiD-CRBN Complex……...……………………………..………………………………….31 5.2 Validation of ZFP91 as a Novel CRBN-CRL4 E3 Ubiquitin Ligase Substrate.………..33 5.3 Effects of ZFP91 Degron Mutagenesis on Substrate Recognition by the iMiD-CRBN Complex…………………………………………..………………….………...34 5.4 iMiD-Induced TNF-! Downregulation in U937 Monocytic Cells…………..……………36 5.5 Effects of CRISPR/Cas9-Mediated CRBN Inactivation on iMiD-Induced TNF-!"Downregulation………………..………………………………………………….….36 6 Discussion…………………………………………………………………………………………38 6.1 Functional Dissection of CRBN-CRL4 Substrate Degrons……..……………………….38 6.2 Investigation of the Molecular Basis for iMiD-Induced TNF-α Downregulation..........41 7 Conclusions ………………………………………………………………………………………44 8 Summary…………………………………………………………………………………………..45 9 Acknowledgments………………………………………………………………………………..46 10 Tables………………………………………………………………………………………………47 11 Figures……………………………………………………………………………………………..50 References……………………………………………………………………………………………….57 2 1 ABSTRACT Background. Thalidomide was first released as a sedative and anti-emetic by a German pharmaceutical company in the 1950s. After its withdrawal from the market due to severe teratogenicity, thalidomide and its novel iMiD analogs were repurposed decades later for a variety of new clinical indications, including the treatment of erythema nodosum leprosum, multiple myeloma, and del(5q)-MDS. Recent work has demonstrated that iMiDs act by modulating the substrate specificity of the CRBN-CRL4 E3 ubiquitin ligase. Selective degradation of IKZF1/IKZF3 and CK1α by this complex in the presence of iMiDs leads to therapeutic activity against multiple myeloma and del(5q)-MDS, respectively. In spite of these advances, our molecular understanding of the pleiotropic effects of iMiDs remains limited. Here, we address two distinct aspects of iMiD biology that warrant further investigation: (1) substrate recognition by the iMiD-CRBN complex via degron motifs and (2) the molecular mechanism of iMiD-induced TNF-α inhibition. Methods. Characterization of iMiD-sensitive degron motifs was performed using a flow cytometry-based fluorescent reporter assay. In brief, HEK293T cells were transfected with GFP- fusion constructs expressing wild-type and mutant degrons of IKZF3 and ZFP91. Substrate degradation in the presence of iMiD treatment was assessed by quantifying GFP reporter expression via flow cytometry. Comparison of a Drug:DMSO GFP ratio between wild-type and mutant degrons was used to determine the effects of single amino acid substitutions on iMiD- induced substrate degradation. The mechanism of TNF-α inhibition by iMiDs was investigated by generating CRISPR/Cas9-mediated single cell knockout clones in the pro-monocytic U937 cell line. Targeted genes were components of the CRBN-CRL4 complex and its associated substrates 3 including CRBN, IKZF1, IKZF3, CSNK1A1, ZFP91, ZNF692, and RNF166. Knockout clones were analyzed by deep sequencing to confirm the presence of inactivating out-of-frame indels. In parallel with non-targeting sgRNA clones, validated knockout clones were treated with PMA to induce macrophagic differentiation, subjected to iMiD treatment, and then stimulated with LPS. TNF-α levels were measured by ELISA to determine any changes resulting from genetic inactivation of candidate targets. Results. Degron mutagenesis studies revealed the presence of critical amino acid residues located in zinc-finger domain 2 of IKZF3 including Q147, C148, G152, A153, and H164. Similarly, amino acids Q401 and G406 in zinc-finger domain 4 were identified as critical residues for the ZFP91 degron. In addition, ZFP91 was validated as a CRBN-CRL4 substrate by Western immunoblotting. Lastly, preliminary TNF-α studies performed to phenotypically characterize CRBN knockout U937 clones suggested a plausible CRBN-dependent mechanism for this iMiD effect, although the results were limited by the presence of clonal heterogeneity. Conclusions. In this study, degron characterization via a fluorescent reporter assay led to the identification of shared amino acid motifs among CRBN-CRL4 substrates that are necessary for conferring sensitivity to iMiD-inducible degradation. Future work employing structural biology approaches would be informative in elucidating the structural determinants of novel zinc-finger substrate recognition by the iMiD-CRBN complex. On the other hand, efforts to dissect the mechanism of iMiD-induced TNF-α inhibition were not fully conclusive. Although the results suggest a plausible CRBN-dependent mechanism, our experimental model warrants further refinement to overcome the limitations encountered in the present study. As an alternative approach, an unbiased proteomics experiment conducted in a monocytic system should be considered to identify novel lineage-specific targets that may provide insight into this elusive mechanism. 4 2 GLOSSARY CK1α……………………………………………..…………………...………………casein kinase 1A1 CRBN……………………………………………………………………………………………...cereblon CRBN-CRL4…………………………….CUL4–RBX1–DDB1–CRBN E3 ubiquitin ligase complex CUL4……………………………………………………………………………….……………….cullin-4 DDB1…………………………………………………………damage-specific DNA-binding protein 1 del(5q)-MDS………………………...….myelodysplastic syndrome with chromosomal 5q deletion ELISA………………………………………...………………….enzyme-linked immunosorbent assay ENL………………………………………..………………...…………....erythema nodosum leprosum FGF2………………………………………..………………………….……...fibroblast growth factor-2 GFP………………………………………………………………………….…green fluorescent protein IFN-γ…………………………………………..…………………………..………….interferon-gamma IKZF1………………………………………..……………………………................................…..Ikaros IKZF3……………………………….………………………………………..……………………….Aiolos IL-2…………………………………………..……………………………...........................interleukin-2 iMiD………………………………………..……………………………... ……immunomodulatory drug LD50……………………………………………………………………………………median lethal dose LPS………………………………………..……………………………................….lipopolysaccharide MM………………………………………..……………………………...................…multiple myeloma NF-!B…………………………………………………………………………….nuclear factor kappa-B NTG……………………………………………...……………………………...….non-targeting sgRNA PBMC…………………………………………..…………………...peripheral blood mononuclear cell RBX1……………………………………………………… ……………………….RING-box protein 1 RNF166………………………………………..………………………..………...ring finger protein 166 TNF-α………………….………………………………………..…………tumor necrosis factor-alpha ZFP91………………………………………..……………………………............zinc-finger protein 91 ZNF692………………………………………..……………………….………...zinc-finger protein 692 VEGF…………………………………………..…………………….vascular endothelial growth factor 5 3 INTRODUCTION 3.1 A TRAGIC BEGINNING Thalidomide is the first member of a class of compounds known as immunomodulatory drugs (iMiDs) that have pleiotropic biological effects including significant anti-neoplastic and anti-inflammatory activities (Table 1) [1,2]. The history of thalidomide as a drug remains exceptionally unique because its legacy has evolved dramatically over the decades since it was first released in 1954 [3]. Introduced as a novel sedative and anti-emetic, thalidomide was initially considered a miracle drug for the treatment of morning sickness in pregnant women. In a tragic turn of events, the drug was banned within a few years due to the widespread emergence of birth defects in children born to mothers who had taken it during their pregnancies [4,5]. Several decades later, thalidomide and its analogs were repurposed as therapeutic agents for the treatment of hematologic malignancies such as multiple myeloma (MM) and del(5q)- myelodysplastic syndrome (MDS) [6,7]. The tortuous history of thalidomide begins with its development by a family-owned pharmaceutical company named Chemie Grünenthal GmbH based in Stolberg, Germany. Established in 1946, this company was the first to introduce penicillin to the German pharmaceutical market in the post-World War II era. To capitalize on its initial success in the antibiotic niche, Chemie
Recommended publications
  • CD29 Identifies IFN-Γ–Producing Human CD8+ T Cells With
    + CD29 identifies IFN-γ–producing human CD8 T cells with an increased cytotoxic potential Benoît P. Nicoleta,b, Aurélie Guislaina,b, Floris P. J. van Alphenc, Raquel Gomez-Eerlandd, Ton N. M. Schumacherd, Maartje van den Biggelaarc,e, and Monika C. Wolkersa,b,1 aDepartment of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands; bLandsteiner Laboratory, Oncode Institute, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; cDepartment of Research Facilities, Sanquin Research, 1066 CX Amsterdam, The Netherlands; dDivision of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; and eDepartment of Molecular and Cellular Haemostasis, Sanquin Research, 1066 CX Amsterdam, The Netherlands Edited by Anjana Rao, La Jolla Institute for Allergy and Immunology, La Jolla, CA, and approved February 12, 2020 (received for review August 12, 2019) Cytotoxic CD8+ T cells can effectively kill target cells by producing therefore developed a protocol that allowed for efficient iso- cytokines, chemokines, and granzymes. Expression of these effector lation of RNA and protein from fluorescence-activated cell molecules is however highly divergent, and tools that identify and sorting (FACS)-sorted fixed T cells after intracellular cytokine + preselect CD8 T cells with a cytotoxic expression profile are lacking. staining. With this top-down approach, we performed an un- + Human CD8 T cells can be divided into IFN-γ– and IL-2–producing biased RNA-sequencing (RNA-seq) and mass spectrometry cells. Unbiased transcriptomics and proteomics analysis on cytokine- γ– – + + (MS) analyses on IFN- and IL-2 producing primary human producing fixed CD8 T cells revealed that IL-2 cells produce helper + + + CD8 Tcells.
    [Show full text]
  • Human Induced Pluripotent Stem Cell–Derived Podocytes Mature Into Vascularized Glomeruli Upon Experimental Transplantation
    BASIC RESEARCH www.jasn.org Human Induced Pluripotent Stem Cell–Derived Podocytes Mature into Vascularized Glomeruli upon Experimental Transplantation † Sazia Sharmin,* Atsuhiro Taguchi,* Yusuke Kaku,* Yasuhiro Yoshimura,* Tomoko Ohmori,* ‡ † ‡ Tetsushi Sakuma, Masashi Mukoyama, Takashi Yamamoto, Hidetake Kurihara,§ and | Ryuichi Nishinakamura* *Department of Kidney Development, Institute of Molecular Embryology and Genetics, and †Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; ‡Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan; §Division of Anatomy, Juntendo University School of Medicine, Tokyo, Japan; and |Japan Science and Technology Agency, CREST, Kumamoto, Japan ABSTRACT Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator–like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in
    [Show full text]
  • ZFP91 (NM 053023) Human Tagged ORF Clone Product Data
    OriGene Technologies, Inc. 9620 Medical Center Drive, Ste 200 Rockville, MD 20850, US Phone: +1-888-267-4436 [email protected] EU: [email protected] CN: [email protected] Product datasheet for RC208217 ZFP91 (NM_053023) Human Tagged ORF Clone Product data: Product Type: Expression Plasmids Product Name: ZFP91 (NM_053023) Human Tagged ORF Clone Tag: Myc-DDK Symbol: ZFP91 Synonyms: DMS-8; DSM-8; DSM8; FKSG11; PZF; ZFP-91; ZNF757 Vector: pCMV6-Entry (PS100001) E. coli Selection: Kanamycin (25 ug/mL) Cell Selection: Neomycin This product is to be used for laboratory only. Not for diagnostic or therapeutic use. View online » ©2021 OriGene Technologies, Inc., 9620 Medical Center Drive, Ste 200, Rockville, MD 20850, US 1 / 5 ZFP91 (NM_053023) Human Tagged ORF Clone – RC208217 ORF Nucleotide >RC208217 ORF sequence Sequence: Red=Cloning site Blue=ORF Green=Tags(s) TTTTGTAATACGACTCACTATAGGGCGGCCGGGAATTCGTCGACTGGATCCGGTACCGAGGAGATCTGCC GCCGCGATCGCC ATGCCGGGGGAGACGGAAGAGCCGAGACCCCCGGAGCAGCAGGACCAGGAAGGGGGAGAGGCGGCCAAGG CGGCTCCGGAGGAGCCCCAACAACGGCCCCCTGAGGCGATCGCGGCGGCGCCTGCAGGGACCACTAGCAG CCGCGTGCTGAGGGGAGGTCGGGACCGAGGCCGGGCCGCTGCGGCCGCCGCCGCCGCAGCTGTGTCCCGC CGGAGGAAGGCCGAGTATCCCCGCCGGCGGAGGAGCAGCCCCAGCGCCAGGCCTCCCGACGTCCCCGGGC AGCAGCCCCAGGCCGCGAAGTCCCCGTCTCCAGTTCAGGGCAAGAAGAGTCCGCGACTCCTATGCATAGA AAAAGTAACAACTGATAAAGATCCCAAGGAAGAAAAAGAGGAAGAAGACGATTCTGCCCTCCCTCAGGAA GTTTCCATTGCTGCATCTAGACCTAGCCGGGGCTGGCGTAGTAGTAGGACATCTGTTTCTCGCCATCGTG ATACAGAGAACACCCGAAGCTCTCGGTCCAAGACCGGTTCATTGCAGCTCATTTGCAAGTCAGAACCAAA TACAGACCAACTTGATTATGATGTTGGAGAAGAGCATCAGTCTCCAGGTGGCATTAGTGAAGAGGAAGAG
    [Show full text]
  • Discovering Pathways in Benign Prostate Hyperplasia: a Functional Genomics Pilot Study
    EXPERIMENTAL AND THERAPEUTIC MEDICINE 21: 242, 2021 Discovering pathways in benign prostate hyperplasia: A functional genomics pilot study ZHELING CHEN1 and MINYAO GE2 1Department of Traditional Chinese Medicine, Zhenxin Community Health Service Center, Shanghai 201824; 2Department of Urology Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China Received February 24, 2020; Accepted October 13, 2020 DOI: 10.3892/etm.2021.9673 Abstract. Benign prostate hyperplasia (BPH) is one of the prevalent in aging males, usually starting at 50‑61 years of well‑known urological neoplasms common in males with an age (1), and causing lower urinary tract symptoms (LUTS), increasing number of associated deaths in aging males. It such as urine flow blockage as a result of the urethra being causes uncomfortable urinary symptoms, including urine flow compressed by the enlarged gland. Other potential complica‑ blockage, and may cause bladder, urinary tract or kidney prob‑ tions may include bladder, urinary tract or kidney problems (2). lems. The histopathological and clinical knowledge regarding Most males have continued prostate growth throughout their BPH is limited. In the present study, an in silico approach was life (2) After the age of 30 years, males exhibit a 1% drop in applied that uses genome‑scale microarray expression data to testosterone production per year and an increase in the level of discover a wide range of protein‑protein interactions in addition dihydrotestosterone, possibly due to the age‑related increase to focusing on specific genes responsible for BPH to develop in 5α reductase (SRD5A2) activity. This rising level of dihy‑ prognostic biomarkers.
    [Show full text]
  • ZFP91—A Newly Described Gene Potentially Involved in Prostate Pathology
    Pathol. Oncol. Res. DOI 10.1007/s12253-013-9716-z RESEARCH ZFP91 —A Newly Described Gene Potentially Involved in Prostate Pathology Lukasz Paschke & Marcin Rucinski & Agnieszka Ziolkowska & Tomasz Zemleduch & Witold Malendowicz & Zbigniew Kwias & Ludwik K. Malendowicz Received: 26 May 2013 /Accepted: 18 October 2013 # The Author(s) 2013. This article is published with open access at Springerlink.com Abstract In search for novel molecular targets in benign pros- Keywords Benign prostate hyperplasia . Prostate cancer . tate hyperplasia (BPH), a PCR Array based screening of 84 Prostate cancer cell lines . ZFP91 genes was performed. Of those, expression of ZFP91 (ZFP91 zinc finger protein) was notably upregulated. Limited data concerning the function of ZFP91 product show that it is a Introduction potential transcription factor upregulated in human acute mye- logenous leukemia and most recently found to be the non- Several lines of evidence show that prostate health could be canonical NF-κB pathway regulator. In order to test this finding adversely affected by obesity [1]. Among other findings, it on a larger number of samples, prostate specimens were obtain- was proven that obesity increases the risk of more aggressive ed from patients undergoing adenomectomy for BPH (n =21), prostate cancer [2], prostate enlargement in men with local- and as a control, from patients undergoing radical cystectomy ized prostate cancer [3], and developing benign prostate hy- for bladder cancer (prostates unchanged pathologically, n =18). perplasia (BPH) [4]. Similar studies were performed on cultured human prostate The starting point for this study was to analyze expression cancer cell lines: LNCaP, DU145, 22Rv1, PC-3; as well as of obesity-related genes in BPH tissue specimens compared to normal prostate epithelial cells—PrEC.
    [Show full text]
  • Cereblon Modulator Iberdomide Induces Degradation of the Transcription Factors Ikaros and Aiolos
    Basic and translational research Ann Rheum Dis: first published as 10.1136/annrheumdis-2017-212916 on 26 June 2018. Downloaded from EXTENDED REPORT Cereblon modulator iberdomide induces degradation of the transcription factors Ikaros and Aiolos: immunomodulation in healthy volunteers and relevance to systemic lupus erythematosus Peter H Schafer,1 Ying Ye,2 Lei Wu,1 Jolanta Kosek,1 Garth Ringheim,1 Zhihong Yang,3 Liangang Liu,3 Michael Thomas,2 Maria Palmisano,2 Rajesh Chopra1,4 Handling editor Josef S ABSTRACT of the autoimmune responses characteristic of Smolen Objectives IKZF1 and IKZF3 (encoding transcription SLE are still being studied, it is clear that Ikaros factors Ikaros and Aiolos) are susceptibility loci for is widely expressed in haematopoietic precursors ► Additional material is published online only. To view systemic lupus erythematosus (SLE). The pharmacology and involved in both lymphoid and myeloid cell please visit the journal online of iberdomide (CC-220), a cereblon (CRBN) modulator development, with Ikaros knockout mice lacking (http:// dx. doi. org/ 10. 1136/ targeting Ikaros and Aiolos, was studied in SLE patient B cells, and Ikaros dominant negative mice lacking annrheumdis- 2017- 212916). cells and in a phase 1 healthy volunteer study. T cells. Aiolos expression is more restricted, found 1Department of Translational Methods CRBN, IKZF1 and IKZF3 gene expression in pre-B cells and mature peripheral B cells, and is Development, Celgene was measured in peripheral blood mononuclear cells required for the generation of long-lived, high-af- Corporation, Summit, New (PBMC) from patients with SLE and healthy volunteers. finity plasma cells.1 Evidence suggests that Ikaros Jersey, USA Ikaros and Aiolos protein levels were measured by regulates key pathways such as the signal trans- 2Department of Clinical Pharmacology, Celgene Western blot and flow cytometry.
    [Show full text]
  • Genome-Wide Loss of Heterozygosity and Copy Number Analysis in Melanoma Using High-Density Single-Nucleotide Polymorphism Arrays Mitchell Stark and Nicholas Hayward
    Research Article Genome-Wide Loss of Heterozygosity and Copy Number Analysis in Melanoma Using High-Density Single-Nucleotide Polymorphism Arrays Mitchell Stark and Nicholas Hayward Oncogenomics Laboratory, Queensland Institute of Medical Research, Herston, Queensland, Australia Abstract Conventional chromosome-based comparative genomic hybridiza- Although a number of genes related to melanoma develop- tion (CGH) has been used to study melanomas of different subtypes ment have been identified through candidate gene screening (4–7), and a limited number of studies have looked at array-based approaches, few studies have attempted to conduct such CGH (aCGH) in murine (8), swine (9), and human (2, 10) mela- analyses on a genome-wide scale. Here we use Illumina 317K nomas. The latter studies have led to the identification of CDK4, whole-genome single-nucleotide polymorphism arrays to CCND1, and KIT amplifications in a subset of malignant melanoma. define a comprehensive allelotype of melanoma based on loss Although aCGH is adequate for detecting high level amplifica- of heterozygosity (LOH) and copy number changes in a panel tions and homozygous deletions (HD), it grossly underestimates of 76 melanoma cell lines. In keeping with previous reports, the level of LOH (11). In contrast, the use of high-density single- we found frequent LOH on chromosome arms 9p (72%), 10p nucleotide polymorphism (SNP) arrays has proved to be a superior approach to defining genome-wide LOH and copy number changes (55%), 10q (55%), 9q (49%), 6q (43%), 11q (43%), and 17p (41%). Tumor suppressor genes (TSGs) can be identified in a wide range of tumor types (e.g., refs.
    [Show full text]
  • An AP-MS- and Bioid-Compatible MAC-Tag Enables Comprehensive Mapping of Protein Interactions and Subcellular Localizations
    ARTICLE DOI: 10.1038/s41467-018-03523-2 OPEN An AP-MS- and BioID-compatible MAC-tag enables comprehensive mapping of protein interactions and subcellular localizations Xiaonan Liu 1,2, Kari Salokas 1,2, Fitsum Tamene1,2,3, Yaming Jiu 1,2, Rigbe G. Weldatsadik1,2,3, Tiina Öhman1,2,3 & Markku Varjosalo 1,2,3 1234567890():,; Protein-protein interactions govern almost all cellular functions. These complex networks of stable and transient associations can be mapped by affinity purification mass spectrometry (AP-MS) and complementary proximity-based labeling methods such as BioID. To exploit the advantages of both strategies, we here design and optimize an integrated approach com- bining AP-MS and BioID in a single construct, which we term MAC-tag. We systematically apply the MAC-tag approach to 18 subcellular and 3 sub-organelle localization markers, generating a molecular context database, which can be used to define a protein’s molecular location. In addition, we show that combining the AP-MS and BioID results makes it possible to obtain interaction distances within a protein complex. Taken together, our integrated strategy enables the comprehensive mapping of the physical and functional interactions of proteins, defining their molecular context and improving our understanding of the cellular interactome. 1 Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland. 2 Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland. 3 Proteomics Unit, University of Helsinki, Helsinki 00014, Finland. Correspondence and requests for materials should be addressed to M.V. (email: markku.varjosalo@helsinki.fi) NATURE COMMUNICATIONS | (2018) 9:1188 | DOI: 10.1038/s41467-018-03523-2 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-03523-2 ajority of proteins do not function in isolation and their proteins.
    [Show full text]
  • IBD Risk Loci Are Enriched in Multigenic Regulatory Modules Encompassing Putative Causative Genes
    ARTICLE DOI: 10.1038/s41467-018-04365-8 OPEN IBD risk loci are enriched in multigenic regulatory modules encompassing putative causative genes Yukihide Momozawa, Julia Dmitrieva et al.# GWAS have identified >200 risk loci for Inflammatory Bowel Disease (IBD). The majority of disease associations are known to be driven by regulatory variants. To identify the putative causative genes that are perturbed by these variants, we generate a large transcriptome data cis 1234567890():,; set (nine disease-relevant cell types) and identify 23,650 -eQTL. We show that these are determined by ∼9720 regulatory modules, of which ∼3000 operate in multiple tissues and ∼970 on multiple genes. We identify regulatory modules that drive the disease association for 63 of the 200 risk loci, and show that these are enriched in multigenic modules. Based on these analyses, we resequence 45 of the corresponding 100 candidate genes in 6600 Crohn disease (CD) cases and 5500 controls, and show with burden tests that they include likely causative genes. Our analyses indicate that ≥10-fold larger sample sizes will be required to demonstrate the causality of individual genes using this approach. Correspondence and requests for materials should be addressed to M.G. (email: [email protected]) #A full list of authors and their affliations appears at the end of the paper. NATURE COMMUNICATIONS | (2018) 9:2427 | DOI: 10.1038/s41467-018-04365-8 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-04365-8 enome Wide Association Studies (GWAS) scan the entire a disease are also associated with changes in expression levels of a genome for statistical associations between common neighboring gene is not sufficient to incriminate the corre- G – variants and disease status in large case control cohorts.
    [Show full text]
  • NF-Kappab-Inducing Kinase in Cancer T ⁎ Gunter Maubach, Michael H
    BBA - Reviews on Cancer 1871 (2019) 40–49 Contents lists available at ScienceDirect BBA - Reviews on Cancer journal homepage: www.elsevier.com/locate/bbacan Review NF-kappaB-inducing kinase in cancer T ⁎ Gunter Maubach, Michael H. Feige, Michelle C.C. Lim, Michael Naumann Institute of Experimental Internal Medicine, Otto von Guericke University, 39120 Magdeburg, Germany ABSTRACT Dysregulation of the alternative NF-κB signaling has severe developmental consequences that can ultimately lead to oncogenesis. Pivotal for the activation of the alternative NF-κB pathway is the stabilization of the NF-κB-inducing kinase (NIK). The aim of this review is to focus on the emerging role of NIK in cancer. The documented subversion of NIK in cancers highlights NIK as a possible therapeutic target. Recent studies show that the alterations of NIK or the components of its regulatory complex are manifold including regulation on the transcript level, copy number changes, mutations as well as protein modifications. High NIK activity is associated with different human malignancies and has adverse effects on tumor patient survival. We discuss here research focusing on deciphering the contribution of NIK towards cancer development and progression. We also report that it is possible to engineer inhibitors with high specificity for NIK and describe developments in this area. 1. Introduction [11], which is phosphorylated by the kinase activity, are bound. The functional relevance of the Thr559 phosphorylation site within The aberrant upregulation of the NF-κB signaling pathway is a the activation loop of the kinase domain (Fig. 1C) however, is still being hallmark of many cancers. In this context, the contribution of the controversially discussed.
    [Show full text]
  • ZFP91 Disturbs Metabolic Fitness and Antitumor Activity of Tumor-Infiltrating T Cells
    The Journal of Clinical Investigation RESEARCH ARTICLE ZFP91 disturbs metabolic fitness and antitumor activity of tumor-infiltrating T cells Feixiang Wang,1 Yuerong Zhang,1 Xiaoyan Yu,1 Xiao-Lu Teng,1 Rui Ding,1 Zhilin Hu,1 Aiting Wang,1 Zhengting Wang,2 Youqiong Ye,1 and Qiang Zou1 1Shanghai Institute of Immunology, Department of Immunology and Microbiology, State Key Laboratory of Oncogenes and Related Genes, Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. 2Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. Proper metabolic activities facilitate T cell expansion and antitumor function; however, the mechanisms underlying disruption of the T cell metabolic program and function in the tumor microenvironment (TME) remain elusive. Here, we show a zinc finger protein 91–governed (ZFP91-governed) mechanism that disrupts the metabolic pathway and antitumor activity of tumor-infiltrating T cells. Single-cell RNA-Seq revealed that impairments in T cell proliferation and activation correlated with ZFP91 in tissue samples from patients with colorectal cancer. T cell–specific deletion of Zfp91 in mice led to enhanced T cell proliferation and potentiated T cell antitumor function. Loss of ZFP91 increased mammalian target of rapamycin complex 1 (mTORC1) activity to drive T cell glycolysis. Mechanistically, T cell antigen receptor–dependent (TCR-dependent) ZFP91 cytosolic translocation promoted protein phosphatase 2A (PP2A) complex assembly, thereby restricting mTORC1-mediated metabolic reprogramming. Our results demonstrate that ZFP91 perturbs T cell metabolic and functional states in the TME and suggest that targeting ZFP91 may improve the efficacy of cancer immunotherapy.
    [Show full text]
  • Avadomide Induces Degradation of ZMYM2 Fusion Oncoproteins in Hematologic Malignancies
    RESEARCH ARTICLE Avadomide Induces Degradation of ZMYM2 Fusion Oncoproteins in Hematologic Malignancies Aline Renneville1,2,3, Jessica A. Gasser1,2, Daniel E. Grinshpun1,2, Pierre M. Jean Beltran1, Namrata D. Udeshi1, Mary E. Matyskiela4, Thomas Clayton4, Marie McConkey2, Kaushik Viswanathan2, Alexander Tepper1,2, Andrew A. Guirguis1,2,5, Rob S. Sellar2,6, Sophie Cotteret7, Christophe Marzac7, Véronique Saada7, Stéphane De Botton8, Jean-Jacques Kiladjian9, Jean-Michel Cayuela10, Mark Rolfe4, Philip P. Chamberlain4, Steven A. Carr1, and Benjamin L. Ebert1,2,11 Downloaded from https://bloodcancerdiscov.aacrjournals.org by guest on September 26, 2021. Copyright 2021 American Copyright 2021 by AssociationAmerican for Association Cancer Research. for Cancer Research. ABSTRACT Thalidomide analogues exert their therapeutic effects by binding to the CRL4CRBN E3 ubiquitin ligase, promoting ubiquitination and subsequent proteasomal degrada- tion of specific protein substrates. Drug-induced degradation of IKZF1 and IKZF3 in B-cell malignancies demonstrates the clinical utility of targeting disease-relevant transcription factors for degradation. Here, we found that avadomide (CC-122) induces CRBN-dependent ubiquitination and proteasomal degradation of ZMYM2 (ZNF198), a transcription factor involved in balanced chromosomal rear- rangements with FGFR1 and FLT3 in aggressive forms of hematologic malignancies. The minimal drug- responsive element of ZMYM2 is a zinc-chelating MYM domain and is contained in the N-terminal portion of ZMYM2 that is universally included in the derived fusion proteins. We demonstrate that avadomide has the ability to induce proteasomal degradation of ZMYM2–FGFR1 and ZMYM2–FLT3 chimeric oncoproteins, both in vitro and in vivo. Our findings suggest that patients with hematologic malignancies harboring these ZMYM2 fusion proteins may benefit from avadomide treatment.
    [Show full text]