1
|
Zhang L, Zhang X, Li Z, Mo T, Feng W, Zhang J, Zhao D, Wang Y, Wei Y, Wang Y. Attenuation of cardiac ischemia/reperfusion injury via the decoy receptor DcR2 by targeting the PLAD domain of the death receptor DR5. Int J Biol Macromol 2025; 308:142529. [PMID: 40154678 DOI: 10.1016/j.ijbiomac.2025.142529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Myocardial cell death caused by ischemia and hypoxia is the main cause of myocardial injury. DcR2 is the decoy receptor of TRAIL, and the role of DcR2 in myocardial ischemia/reperfusion (I/R) injury is largely unknown. Recent studies have shown that DcR2 not only binds to TRAIL as a receptor but also acts as a ligand for DR5 to block TRAIL-induced apoptosis in vitro, but the preference of DcR2 for binding to TRAIL or DR5 in vivo remains unknown. Our study revealed that the hDcR2-Fc fusion protein plays a cardioprotective role in a mouse model of myocardial I/R injury by reducing apoptosis. An affinity assay revealed that DcR2 has a greater affinity for DR5 than for TRAIL and that DcR2 is more inclined to bind to DR5. Mechanistic studies elucidated that deletion of PLAD eliminated the protective effect of hDcR2-Fc on heart injury caused by I/R. DcR2 forms a heterocomplex with DR5 through a similar PLAD domain. Taken together, this study revealed that DcR2 can ameliorate myocardial I/R injury by targeting DR5 to form a heterocomplex through the PLAD domain, blocking apoptosis, thus providing a new preventive strategy for the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Lijie Zhang
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, China; Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Xinyuan Zhang
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, China; Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Ziting Li
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, China
| | - Tingting Mo
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, China
| | - Wanting Feng
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, China
| | - JingLun Zhang
- School of Medicine, Henan University, Kaifeng, China
| | - Dan Zhao
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, China
| | - Ying Wang
- School of Medicine, Henan University, Kaifeng, China
| | - Yinxiang Wei
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, China.
| | - Yaohui Wang
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, China.
| |
Collapse
|
2
|
Micheau O, Fournel S. Comment on "Self-Illuminating Nanoagonist Simultaneously Induces Dual Cell Death Pathways via Death Receptor Clustering for Cancer Therapy". ACS NANO 2025; 19:1-3. [PMID: 39810374 DOI: 10.1021/acsnano.4c13100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Affiliation(s)
- Olivier Micheau
- Université de Bourgogne, U1231 INSERM, CTM, Equipe DesCarTes, UFR Sciences de Santé, 7 Bd Jeanne d'Arc, 21078 Dijon Cedex, France
| | - Sylvie Fournel
- Université de Strasbourg UMR _S 1121, EMR CNRS 7003, Biomatériaux & Bioingénierie, CRBS, Centre de Recherche en Biomédecine de Strasbourg, 1 Rue Eugène Boeckel, 67084 Strasbourg Cedex, France
| |
Collapse
|
3
|
Xing L, Jiang Z, Xu R, Dang T, Wu J, Chai J, Meng X. CCN1 promotes APRIL/BAFF signaling in esophageal squamous cell carcinoma but attenuates it in esophageal adenocarcinoma. Sci Rep 2025; 15:1808. [PMID: 39806221 PMCID: PMC11730293 DOI: 10.1038/s41598-025-86228-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025] Open
Abstract
CCN1 is a matricellular protein highly expressed in esophageal squamous cell carcinoma (ESCC) but hardly detectable in esophageal adenocarcinoma (EAC). Expression of CCN1 in EAC cells leads to TRAIL-mediated apoptosis. Unlike TRAIL, which primarily triggers cell death, APRIL and BAFF promote cell growth via NFκB signaling. They become active ligands by Furin cleavage. This study found that CCN1 upregulated APRIL and BAFF expression in both ESCC and EAC cells but attenuated their signaling in the latter. CCN1 kept Furin stable in ESCC allowing APRIL/BAFF to signal through their common receptor BCMA properly. In EAC cells, however, expression of CCN1 lowered Furin activity and thus limited APRIL/BAFF cleavage. As a result, ESCC cells benefited from CCN1 while EAC cell viability was attenuated by it.
Collapse
Affiliation(s)
- Lingling Xing
- Inner Mongolia Institute of Digestive Diseases, Baotou, China
- Inner Mongolia Engineering Research Center for Prevention and Treatment of Digestive Diseases, Baotou, China
- The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, 30 Hudemulin Rd, Baotou, 014030, China
| | - Zhenyu Jiang
- Inner Mongolia Institute of Digestive Diseases, Baotou, China
- Inner Mongolia Engineering Research Center for Prevention and Treatment of Digestive Diseases, Baotou, China
- The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, 30 Hudemulin Rd, Baotou, 014030, China
| | - Ruize Xu
- Inner Mongolia Institute of Digestive Diseases, Baotou, China
- Inner Mongolia Engineering Research Center for Prevention and Treatment of Digestive Diseases, Baotou, China
- The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, 30 Hudemulin Rd, Baotou, 014030, China
| | - Tong Dang
- Inner Mongolia Institute of Digestive Diseases, Baotou, China
- Inner Mongolia Engineering Research Center for Prevention and Treatment of Digestive Diseases, Baotou, China
- The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, 30 Hudemulin Rd, Baotou, 014030, China
| | - Jinbao Wu
- Inner Mongolia Institute of Digestive Diseases, Baotou, China.
- Inner Mongolia Engineering Research Center for Prevention and Treatment of Digestive Diseases, Baotou, China.
- The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, 30 Hudemulin Rd, Baotou, 014030, China.
| | - Jianyuan Chai
- Inner Mongolia Institute of Digestive Diseases, Baotou, China.
- Inner Mongolia Engineering Research Center for Prevention and Treatment of Digestive Diseases, Baotou, China.
- The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, 30 Hudemulin Rd, Baotou, 014030, China.
| | - Xianmei Meng
- Inner Mongolia Institute of Digestive Diseases, Baotou, China.
- Inner Mongolia Engineering Research Center for Prevention and Treatment of Digestive Diseases, Baotou, China.
- The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, 30 Hudemulin Rd, Baotou, 014030, China.
| |
Collapse
|
4
|
Batiste M, Joy B, Yee CK, Cho L, Christensen A, Abed I, Nguyen K, Yanumula A, Chang H, Cho ED, Wang W, Chou E, Chang EH, Shyu YL, Abram A, Alcaide J, Zhou J, Gillespie B, Senderovich M, Cusick GA, Le AV, Hoang F, Shi Y, Mohamed E, Cusick JK. RELT Is Upregulated in Breast Cancer and Induces Death in Breast Cancer Cells. Biomedicines 2024; 12:2667. [PMID: 39767574 PMCID: PMC11727564 DOI: 10.3390/biomedicines12122667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/02/2024] [Accepted: 11/08/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Receptor Expressed in Lymphoid Tissues (RELT) is a TNFRSF member that has two paralogs, RELL1 and RELL2; the three proteins are collectively referred to as RELT family members (RELTfms). METHODS We sought to evaluate RELT expression in cancerous cells by using real-time PCR, western blotting, flow cytometry, and immunohistochemistry (IHC). The mechanism of RELT-induced cell death was assessed by western blotting, flow cytometry, luciferase assays, and morphology staining. RELT localization was detected through immunofluorescence and western blotting, and co-immunoprecipitation was used to test whether a mutated RELT interacts with the OXSR1 kinase. RESULTS RELT and RELL1 protein expression was significantly elevated in cell lines representing breast and lung cancer, whereas RELL2 protein expression was relatively consistent across different cell lines. The surface expression of RELT was highest in monocytes. IHC staining revealed increased RELT expression in malignant breast cancer biopsies compared to patient-matched benign tissue. RELTfm overexpression induced death in MDA-MB-231 (231) breast cancer cells, accompanied by increased phosphatidylserine externalization and Caspase-3/7 activation. The co-transfection of plasmids predicted to block the phosphorylation of RELT by the OXSR1 kinase did not abrogate RELT-induced apoptosis, indicating that the activation of p38 by RELT through the OXSR1 kinase is not required for RELT-induced cell death. Interestingly, nuclear localization of RELT was detected in 231 and HEK-293 cells. CONCLUSIONS These results demonstrate that RELT induces death in breast cancer cells through an apoptotic pathway that does not require OXSR1 phosphorylation and that RELT possesses the ability to translocate to the nucleus, a novel finding that warrants further investigation.
Collapse
Affiliation(s)
- Maryann Batiste
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Bethany Joy
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Cara K. Yee
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Luke Cho
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Ashley Christensen
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Ihab Abed
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Kailey Nguyen
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Anusri Yanumula
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Hannah Chang
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Evan D. Cho
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Wenjia Wang
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Emily Chou
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Esther H. Chang
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Yennie L. Shyu
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Alyssa Abram
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Jessa Alcaide
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - James Zhou
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Brittany Gillespie
- Masters of Pharmaceutical Sciences Department, College of Graduate Studies, California Northstate University, Elk Grove, CA 95757, USA
| | - Michelle Senderovich
- Masters of Pharmaceutical Sciences Department, College of Graduate Studies, California Northstate University, Elk Grove, CA 95757, USA
| | - Gianne Almeida Cusick
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Ai-Vy Le
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Frank Hoang
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| | - Yihui Shi
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
- California Pacific Medical Center Research Institute, San Francisco, CA 94107, USA
| | - Eslam Mohamed
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
- Masters of Pharmaceutical Sciences Department, College of Graduate Studies, California Northstate University, Elk Grove, CA 95757, USA
| | - John K. Cusick
- Department of Basic Science, College of Medicine, California Northstate University, Elk Grove, CA 95757, USA (B.J.); (A.C.); (H.C.); (E.D.C.); (E.H.C.); (Y.L.S.); (A.-V.L.); (Y.S.); (E.M.)
| |
Collapse
|
5
|
Chiou S, Cawthorne W, Soerianto T, Hofferek V, Patel KM, Garnish SE, Tovey Crutchfield EC, Hall C, Hildebrand JM, McConville MJ, Lawlor KE, Hawkins ED, Samson AL, Murphy JM. MLKL deficiency elevates testosterone production in male mice independently of necroptotic functions. Cell Death Dis 2024; 15:851. [PMID: 39572538 PMCID: PMC11582601 DOI: 10.1038/s41419-024-07242-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024]
Abstract
Mixed lineage kinase domain-like (MLKL) is a pseudokinase, best known for its role as the terminal effector of the necroptotic cell death pathway. MLKL-mediated necroptosis has long been linked to various age-related pathologies including neurodegeneration, atherosclerosis and male reproductive decline, however many of these attributions remain controversial. Here, we investigated the role of MLKL and necroptosis in the adult mouse testis: an organ divided into sperm-producing seminiferous tubules and the surrounding testosterone-producing interstitium. We find that sperm-producing cells within seminiferous tubules lack expression of key necroptotic mediators and thus are resistant to a pro-necroptotic challenge. By comparison, coordinated expression of the necroptotic pathway occurs in the testicular interstitium, rendering cells within this compartment, especially the lysozyme-positive macrophages, vulnerable to necroptotic cell death. We also uncover a non-necroptotic role for MLKL in regulating testosterone levels. Thus, MLKL serves two roles in the mouse testes - one involving the canonical response of macrophages to necroptotic insult, and the other a non-canonical function in male reproductive hormone control.
Collapse
Affiliation(s)
- Shene Chiou
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Wayne Cawthorne
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Thomas Soerianto
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia
| | - Vinzenz Hofferek
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia
| | - Komal M Patel
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Sarah E Garnish
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Emma C Tovey Crutchfield
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Department of Ophthalmology, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Cathrine Hall
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Joanne M Hildebrand
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Malcolm J McConville
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia
| | - Kate E Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Edwin D Hawkins
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - Andre L Samson
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
| |
Collapse
|
6
|
Norris MJ, Henderson LA, Siddiquey MNA, Yin J, Yoo K, Brunel S, Saphire EO, Benedict CA, Kamil JP. A noncanonical glycoprotein H complex enhances cytomegalovirus entry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.617647. [PMID: 39416215 PMCID: PMC11482907 DOI: 10.1101/2024.10.13.617647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Human cytomegalovirus (HCMV) causes severe birth defects, lifelong health complications, and $4 billion in annual costs in the United States alone. A major challenge in vaccine design is the incomplete understanding of the diverse protein complexes the virus uses to infect cells. In Herpesviridae, the gH/gL glycoprotein heterodimer is expected to be a basal element of virion cell entry machinery. For HCMV, gH/gL forms a "trimer" with gO and a "pentamer" with UL128, UL130, and UL131A, with each complex binding distinct receptors to enter varied cell types. Here, we reveal a third glycoprotein complex, abundant in HCMV virions, which significantly enhances infection of endothelial cells. In this "3-mer" complex, gH, without gL, associates with UL116 and UL141, an immunoevasin previously known to function in an intracellular role. Cryo-EM reveals the virion-surface 3-mer is structurally unique among Herpesviridae gH complexes, with gH-only scaffolding, UL141-mediated dimerization and a heavily glycosylated UL116 cap. Given that antibodies directed at gH and UL141 each can restrict HCMV replication, our work highlights this virion surface complex as a new target for vaccines and antiviral therapies.
Collapse
Affiliation(s)
- Michael J Norris
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA
- Present address (M.N.): Department of Biochemistry, University of Toronto, Toronto, ON Canada
| | - Lauren A Henderson
- Department of Microbiology and Immunology, Louisiana State University Health Shreveport, Shreveport, Shreveport, LA
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Shreveport, Shreveport, LA
| | - Mohammed N A Siddiquey
- Department of Microbiology and Immunology, Louisiana State University Health Shreveport, Shreveport, Shreveport, LA
| | - Jieyun Yin
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA
| | - Kwangsun Yoo
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA
| | - Simon Brunel
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Chris A Benedict
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA
| | - Jeremy P Kamil
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
7
|
Garnish SE, Horne CR, Meng Y, Young SN, Jacobsen AV, Hildebrand JM, Murphy JM. Inhibitors identify an auxiliary role for mTOR signalling in necroptosis execution downstream of MLKL activation. Biochem J 2024; 481:1125-1142. [PMID: 39136677 PMCID: PMC11555701 DOI: 10.1042/bcj20240255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
Necroptosis is a lytic and pro-inflammatory form of programmed cell death executed by the terminal effector, the MLKL (mixed lineage kinase domain-like) pseudokinase. Downstream of death and Toll-like receptor stimulation, MLKL is trafficked to the plasma membrane via the Golgi-, actin- and microtubule-machinery, where activated MLKL accumulates until a critical lytic threshold is exceeded and cell death ensues. Mechanistically, MLKL's lytic function relies on disengagement of the N-terminal membrane-permeabilising four-helix bundle domain from the central autoinhibitory brace helix: a process that can be experimentally mimicked by introducing the R30E MLKL mutation to induce stimulus-independent cell death. Here, we screened a library of 429 kinase inhibitors for their capacity to block R30E MLKL-mediated cell death, to identify co-effectors in the terminal steps of necroptotic signalling. We identified 13 compounds - ABT-578, AR-A014418, AZD1480, AZD5363, Idelalisib, Ipatasertib, LJI308, PHA-793887, Rapamycin, Ridaforolimus, SMI-4a, Temsirolimus and Tideglusib - each of which inhibits mammalian target of rapamycin (mTOR) signalling or regulators thereof, and blocked constitutive cell death executed by R30E MLKL. Our study implicates mTOR signalling as an auxiliary factor in promoting the transport of activated MLKL oligomers to the plasma membrane, where they accumulate into hotspots that permeabilise the lipid bilayer to cause cell death.
Collapse
Affiliation(s)
- Sarah E. Garnish
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Christopher R. Horne
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Yanxiang Meng
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Samuel N. Young
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
| | - Annette V. Jacobsen
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Joanne M. Hildebrand
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - James M. Murphy
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
8
|
Li W, Xia C, Wang K, Xue L, Wang Y, Yang JY, Zhang M, Yin M, Ju C, Miao Z, Li Y, Zhao X, Yang Z, Tang R, Yang W. Technical considerations and strategies for generating and optimizing humanized mouse tumor models in immuno-oncology research. Int Immunopharmacol 2024; 139:112722. [PMID: 39033663 DOI: 10.1016/j.intimp.2024.112722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
The field of cancer immunotherapy has experienced significant progress, resulting in the emergence of numerous biological drug candidates requiring in vivo efficacy testing and a better understanding of their mechanism of action (MOA). Humanized immune system (HIS) models are valuable tools in this regard. However, there is a lack of systematic guidance on HIS modeling. To address this issue, the present study aimed to establish and optimize a variety of HIS models for immune-oncology (IO) study, including genetically engineered mouse models and HIS models with human immune components reconstituted in severely immunocompromised mice. The efficacy and utility of these models were tested with several marketed or investigational IO drugs according to their MOA, followed by immunophenotypic analysis and efficacy evaluation. The results of the present study demonstrated that the HIS models responded to various IO drugs as expected and that each model had unique niches, utilities and limitations. Researchers should carefully choose the appropriate models based on the MOA and the targeted immune cell populations of the investigational drug. The present study provides valuable methodologies and actionable technical guidance on designing, generating or utilizing appropriate HIS models to address specific questions in translational IO.
Collapse
Affiliation(s)
- Wenjing Li
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China
| | - Chunlei Xia
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China
| | - Kun Wang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China
| | - Liting Xue
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China
| | - Yan Wang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China
| | | | | | - Ming Yin
- Beijing Vitalstar Biotechnology Co., Ltd., Beijing 100000, China
| | - Cunxiang Ju
- Gempharmatech Co., Ltd., Nanjing 210000, China
| | - Zhenchuan Miao
- Beijing Vitalstar Biotechnology Co., Ltd., Beijing 100000, China
| | - Ying Li
- Gempharmatech Co., Ltd., Nanjing 210000, China
| | - Xiaofeng Zhao
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Jiangsu Simcere Pharmaceutical Co, Ltd., Nanjing 210000, China
| | - Zhijian Yang
- ClinBridge Biotech Co., Ltd., Nanjing 210000, China
| | - Renhong Tang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China.
| | - WenQing Yang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, China; Simcere Zaiming Pharmaceutical Co, Ltd., Shanghai 200120, China.
| |
Collapse
|
9
|
Sim N, Carter JM, Deka K, Tan BKT, Sim Y, Tan SM, Li Y. TWEAK/Fn14 signalling driven super-enhancer reprogramming promotes pro-metastatic metabolic rewiring in triple-negative breast cancer. Nat Commun 2024; 15:5638. [PMID: 38965263 PMCID: PMC11224303 DOI: 10.1038/s41467-024-50071-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 06/27/2024] [Indexed: 07/06/2024] Open
Abstract
Triple Negative Breast Cancer (TNBC) is the most aggressive breast cancer subtype suffering from limited targeted treatment options. Following recent reports correlating Fibroblast growth factor-inducible 14 (Fn14) receptor overexpression in Estrogen Receptor (ER)-negative breast cancers with metastatic events, we show that Fn14 is specifically overexpressed in TNBC patients and associated with poor survival. We demonstrate that constitutive Fn14 signalling rewires the transcriptomic and epigenomic landscape of TNBC, leading to enhanced tumour growth and metastasis. We further illustrate that such mechanisms activate TNBC-specific super enhancers (SE) to drive the transcriptional activation of cancer dependency genes via chromatin looping. In particular, we uncover the SE-driven upregulation of Nicotinamide phosphoribosyltransferase (NAMPT), which promotes NAD+ and ATP metabolic reprogramming critical for filopodia formation and metastasis. Collectively, our study details the complex mechanistic link between TWEAK/Fn14 signalling and TNBC metastasis, which reveals several vulnerabilities which could be pursued for the targeted treatment of TNBC patients.
Collapse
Affiliation(s)
- Nicholas Sim
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Jean-Michel Carter
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Kamalakshi Deka
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Benita Kiat Tee Tan
- Division of Surgery and Surgical Oncology, Department of Breast Surgery, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore, 168583, Singapore
- Division of Surgery and Surgical Oncology, Department of Breast Surgery, Singapore General Hospital, 31 Third Hospital Ave, Singapore, 168753, Singapore
- SingHealth Duke-NUS Breast Centre, Singapore, Singapore
| | - Yirong Sim
- Division of Surgery and Surgical Oncology, Department of Breast Surgery, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore, 168583, Singapore
- Division of Surgery and Surgical Oncology, Department of Breast Surgery, Singapore General Hospital, 31 Third Hospital Ave, Singapore, 168753, Singapore
- SingHealth Duke-NUS Breast Centre, Singapore, Singapore
| | - Suet-Mien Tan
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Yinghui Li
- School of Biological Sciences (SBS), Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore, 637551, Singapore.
| |
Collapse
|
10
|
Chiou S, Al-Ani AH, Pan Y, Patel KM, Kong IY, Whitehead LW, Light A, Young SN, Barrios M, Sargeant C, Rajasekhar P, Zhu L, Hempel A, Lin A, Rickard JA, Hall C, Gangatirkar P, Yip RK, Cawthorne W, Jacobsen AV, Horne CR, Martin KR, Ioannidis LJ, Hansen DS, Day J, Wicks IP, Law C, Ritchie ME, Bowden R, Hildebrand JM, O'Reilly LA, Silke J, Giulino-Roth L, Tsui E, Rogers KL, Hawkins ED, Christensen B, Murphy JM, Samson AL. An immunohistochemical atlas of necroptotic pathway expression. EMBO Mol Med 2024; 16:1717-1749. [PMID: 38750308 PMCID: PMC11250867 DOI: 10.1038/s44321-024-00074-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 06/12/2024] Open
Abstract
Necroptosis is a lytic form of regulated cell death reported to contribute to inflammatory diseases of the gut, skin and lung, as well as ischemic-reperfusion injuries of the kidney, heart and brain. However, precise identification of the cells and tissues that undergo necroptotic cell death in vivo has proven challenging in the absence of robust protocols for immunohistochemical detection. Here, we provide automated immunohistochemistry protocols to detect core necroptosis regulators - Caspase-8, RIPK1, RIPK3 and MLKL - in formalin-fixed mouse and human tissues. We observed surprising heterogeneity in protein expression within tissues, whereby short-lived immune barrier cells were replete with necroptotic effectors, whereas long-lived cells lacked RIPK3 or MLKL expression. Local changes in the expression of necroptotic effectors occurred in response to insults such as inflammation, dysbiosis or immune challenge, consistent with necroptosis being dysregulated in disease contexts. These methods will facilitate the precise localisation and evaluation of necroptotic signaling in vivo.
Collapse
Affiliation(s)
- Shene Chiou
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Aysha H Al-Ani
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
- Royal Melbourne Hospital, Parkville, Australia
| | - Yi Pan
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Komal M Patel
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Isabella Y Kong
- Pediatric Hematology/Oncology, Weill Cornell Medical College, New York, USA
| | - Lachlan W Whitehead
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Amanda Light
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Samuel N Young
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Marilou Barrios
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Callum Sargeant
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Pradeep Rajasekhar
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Leah Zhu
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Anne Hempel
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Ann Lin
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - James A Rickard
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Austin Hospital, Heidelberg, Australia
| | - Cathrine Hall
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | | | - Raymond Kh Yip
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Wayne Cawthorne
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Annette V Jacobsen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Christopher R Horne
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Katherine R Martin
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Lisa J Ioannidis
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Diana S Hansen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Australia
| | - Jessica Day
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
- Royal Melbourne Hospital, Parkville, Australia
| | - Ian P Wicks
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Charity Law
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Matthew E Ritchie
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Rory Bowden
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Joanne M Hildebrand
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Lorraine A O'Reilly
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - John Silke
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Lisa Giulino-Roth
- Pediatric Hematology/Oncology, Weill Cornell Medical College, New York, USA
| | - Ellen Tsui
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Kelly L Rogers
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Edwin D Hawkins
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
| | - Britt Christensen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- University of Melbourne, Parkville, Australia
- Royal Melbourne Hospital, Parkville, Australia
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.
- University of Melbourne, Parkville, Australia.
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia.
| | - André L Samson
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.
- University of Melbourne, Parkville, Australia.
| |
Collapse
|
11
|
Wang Y, Wang E, Anany M, Füllsack S, Huo YH, Dutta S, Ji B, Hoeppner LH, Kilari S, Misra S, Caulfield T, Vander Kooi CW, Wajant H, Mukhopadhyay D. The crosstalk between neuropilin-1 and tumor necrosis factor-α in endothelial cells. Front Cell Dev Biol 2024; 12:1210944. [PMID: 38994453 PMCID: PMC11236538 DOI: 10.3389/fcell.2024.1210944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
Tumor necrosis factor-α (TNFα) is a master cytokine which induces expression of chemokines and adhesion molecules, such as intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1), in endothelial cells to initiate the vascular inflammatory response. In this study, we identified neuropilin-1 (NRP1), a co-receptor of several structurally diverse ligands, as a modulator of TNFα-induced inflammatory response of endothelial cells. NRP1 shRNA expression suppressed TNFα-stimulated leukocyte adhesion and expression of ICAM-1 and VCAM-1 in human umbilical vein endothelial cells (HUVECs). Likewise, it reduced TNFα-induced phosphorylation of MAPK p38 but did not significantly affect other TNF-induced signaling pathways, such as the classical NFκB and the AKT pathway. Immunofluorescent staining demonstrated co-localization of NRP1 with the two receptors of TNF, TNFR1 and TNFR2. Co-immunoprecipitation further confirmed that NRP1 was in the same protein complex or membrane compartment as TNFR1 and TNFR2, respectively. Modulation of NRP1 expression, however, neither affected TNFR levels in the cell membrane nor the receptor binding affinities of TNFα. Although a direct interface between NRP1 and TNFα/TNFR1 appeared possible from a protein docking model, a direct interaction was not supported by binding assays in cell-free microplates and cultured cells. Furthermore, TNFα was shown to downregulate NRP1 in a time-dependent manner through TNFR1-NFκB pathway in HUVECs. Taken together, our study reveals a novel reciprocal crosstalk between NRP1 and TNFα in vascular endothelial cells.
Collapse
Affiliation(s)
- Ying Wang
- Department of Cardiovascular Medicine, Rochester, MN, United States
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, United States
| | - Mohamed Anany
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- Department of Microbial Biotechnology, Institute of Biotechnology, National Research Centre, Giza, Egypt
| | - Simone Füllsack
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Yu Henry Huo
- Department of Cardiovascular Medicine, Rochester, MN, United States
| | - Shamit Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, United States
| | - Baoan Ji
- Department of Cancer Biology, Jacksonville, FL, United States
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | | | - Sanjay Misra
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Thomas Caulfield
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Craig W Vander Kooi
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, United States
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
12
|
Day ZI, Mayfosh AJ, Baxter AA, Williams SA, Hildebrand JM, Rau TF, Poon IKH, Hulett MD. Defining a Water-Soluble Formulation of Arachidonic Acid as a Novel Ferroptosis Inducer in Cancer Cells. Biomolecules 2024; 14:555. [PMID: 38785962 PMCID: PMC11118058 DOI: 10.3390/biom14050555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Here, we describe GS-9, a novel water-soluble fatty acid-based formulation comprising L-lysine and arachidonic acid, that we have shown to induce ferroptosis. GS-9 forms vesicle-like structures in solution and mediates lipid peroxidation, as evidenced by increased C11-BODIPY fluorescence and an accumulation of toxic malondialdehyde, a downstream product of lipid peroxidation. Ferroptosis inhibitors counteracted GS-9-induced cell death, whereas caspase 3 and 7 or MLKL knock-out cell lines are resistant to GS-9-induced cell death, eliminating other cell death processes such as apoptosis and necroptosis as the mechanism of action of GS-9. We also demonstrate that through their role of sequestering fatty acids, lipid droplets play a protective role against GS-9-induced ferroptosis, as inhibition of lipid droplet biogenesis enhanced GS-9 cytotoxicity. In addition, Fatty Acid Transport Protein 2 was implicated in GS-9 uptake. Overall, this study identifies and characterises the mechanism of GS-9 as a ferroptosis inducer. This formulation of arachidonic acid offers a novel tool for investigating and manipulating ferroptosis in various cellular and anti-cancer contexts.
Collapse
Affiliation(s)
- Zoe I. Day
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (Z.I.D.); (A.J.M.); (A.A.B.); (I.K.H.P.)
| | - Alyce J. Mayfosh
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (Z.I.D.); (A.J.M.); (A.A.B.); (I.K.H.P.)
- Wintermute Biomedical, Geelong, VIC 3220, Australia
| | - Amy A. Baxter
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (Z.I.D.); (A.J.M.); (A.A.B.); (I.K.H.P.)
| | - Scott A. Williams
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (Z.I.D.); (A.J.M.); (A.A.B.); (I.K.H.P.)
| | - Joanne M. Hildebrand
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia;
| | | | - Ivan K. H. Poon
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (Z.I.D.); (A.J.M.); (A.A.B.); (I.K.H.P.)
| | - Mark D. Hulett
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (Z.I.D.); (A.J.M.); (A.A.B.); (I.K.H.P.)
| |
Collapse
|
13
|
Kim AS, Taylor VE, Castro-Martinez A, Dhakal S, Zamerli A, Mohanty S, Xiao Y, Simic MK, Wen J, Chai R, Croucher PI, Center JR, Girgis CM, McDonald MM. Temporal patterns of osteoclast formation and activity following withdrawal of RANKL inhibition. J Bone Miner Res 2024; 39:484-497. [PMID: 38477789 PMCID: PMC11262142 DOI: 10.1093/jbmr/zjae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 01/15/2024] [Accepted: 01/25/2024] [Indexed: 03/14/2024]
Abstract
Rebound bone loss following denosumab discontinuation is an important clinical challenge. Current treatment strategies to prevent this fail to suppress the rise and overshoot in osteoclast-mediated bone resorption. In this study, we use a murine model of denosumab treatment and discontinuation to show the temporal changes in osteoclast formation and activity during RANKL inhibition and withdrawal. We show that the cellular processes that drive the formation of osteoclasts and subsequent bone resorption following withdrawal of RANKL inhibition precede the rebound bone loss. Furthermore, a rise in serum TRAP and RANKL levels is detected before markers of bone turnover used in current clinical practice. These mechanistic advances may provide insight into a more defined window of opportunity to intervene with sequential therapy following denosumab discontinuation.
Collapse
Affiliation(s)
- Albert S Kim
- Skeletal Diseases Program, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
- Faculty of Medicine, St Vincent’s Clinical School, UNSW Sydney, Sydney, NSW, 2010, Australia
- Department of Diabetes and Endocrinology, Westmead Hospital, Sydney, NSW, 2145, Australia
- Faculty of Health and Medicine, University of Sydney, Sydney, NSW, 2050, Australia
| | - Victoria E Taylor
- Skeletal Diseases Program, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Ariel Castro-Martinez
- Skeletal Diseases Program, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Suraj Dhakal
- Skeletal Diseases Program, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Amjad Zamerli
- Skeletal Diseases Program, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Sindhu Mohanty
- Skeletal Diseases Program, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Ya Xiao
- Skeletal Diseases Program, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Marija K Simic
- Skeletal Diseases Program, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, United States
| | - Jinchen Wen
- Department of Psychology and Neuroscience, Duke University, Durham, NC, 27708, United States
| | - Ryan Chai
- Skeletal Diseases Program, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
- Faculty of Medicine, St Vincent’s Clinical School, UNSW Sydney, Sydney, NSW, 2010, Australia
| | - Peter I Croucher
- Skeletal Diseases Program, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
- Faculty of Medicine, St Vincent’s Clinical School, UNSW Sydney, Sydney, NSW, 2010, Australia
| | - Jacqueline R Center
- Skeletal Diseases Program, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
- Faculty of Medicine, St Vincent’s Clinical School, UNSW Sydney, Sydney, NSW, 2010, Australia
| | - Christian M Girgis
- Department of Diabetes and Endocrinology, Westmead Hospital, Sydney, NSW, 2145, Australia
- Faculty of Health and Medicine, University of Sydney, Sydney, NSW, 2050, Australia
| | - Michelle M McDonald
- Skeletal Diseases Program, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
- Faculty of Medicine, St Vincent’s Clinical School, UNSW Sydney, Sydney, NSW, 2010, Australia
- Faculty of Health and Medicine, University of Sydney, Sydney, NSW, 2050, Australia
| |
Collapse
|
14
|
Mischley V, Maier J, Chen J, Karanicolas J. PPIscreenML: Structure-based screening for protein-protein interactions using AlphaFold. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.16.585347. [PMID: 38559274 PMCID: PMC10979958 DOI: 10.1101/2024.03.16.585347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Protein-protein interactions underlie nearly all cellular processes. With the advent of protein structure prediction methods such as AlphaFold2 (AF2), models of specific protein pairs can be built extremely accurately in most cases. However, determining the relevance of a given protein pair remains an open question. It is presently unclear how to use best structure-based tools to infer whether a pair of candidate proteins indeed interact with one another: ideally, one might even use such information to screen amongst candidate pairings to build up protein interaction networks. Whereas methods for evaluating quality of modeled protein complexes have been co-opted for determining which pairings interact (e.g., pDockQ and iPTM), there have been no rigorously benchmarked methods for this task. Here we introduce PPIscreenML, a classification model trained to distinguish AF2 models of interacting protein pairs from AF2 models of compelling decoy pairings. We find that PPIscreenML out-performs methods such as pDockQ and iPTM for this task, and further that PPIscreenML exhibits impressive performance when identifying which ligand/receptor pairings engage one another across the structurally conserved tumor necrosis factor superfamily (TNFSF). Analysis of benchmark results using complexes not seen in PPIscreenML development strongly suggest that the model generalizes beyond training data, making it broadly applicable for identifying new protein complexes based on structural models built with AF2.
Collapse
Affiliation(s)
- Victoria Mischley
- Cancer Signaling & Microenvironment Program, Fox Chase Cancer Center, Philadelphia PA 19111
- Molecular Cell Biology and Genetics, Drexel University, Philadelphia PA 19102
| | | | | | - John Karanicolas
- Cancer Signaling & Microenvironment Program, Fox Chase Cancer Center, Philadelphia PA 19111
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia PA 19140
| |
Collapse
|
15
|
Liu Y, Zhang Y, Chang X, Liu X. MDIC3: Matrix decomposition to infer cell-cell communication. PATTERNS (NEW YORK, N.Y.) 2024; 5:100911. [PMID: 38370122 PMCID: PMC10873161 DOI: 10.1016/j.patter.2023.100911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/31/2023] [Accepted: 12/08/2023] [Indexed: 02/20/2024]
Abstract
Crosstalk among cells is vital for maintaining the biological function and intactness of systems. Most existing methods for investigating cell-cell communications are based on ligand-receptor (L-R) expression, and they focus on the study between two cells. Thus, the final communication inference results are particularly sensitive to the completeness and accuracy of the prior biological knowledge. Because existing L-R research focuses mainly on humans, most existing methods can only examine cell-cell communication for humans. As far as we know, there is currently no effective method to overcome this species limitation. Here, we propose MDIC3 (matrix decomposition to infer cell-cell communication), an unsupervised tool to investigate cell-cell communication in any species, and the results are not limited by specific L-R pairs or signaling pathways. By comparing it with existing methods for the inference of cell-cell communication, MDIC3 obtained better performance in both humans and mice.
Collapse
Affiliation(s)
- Yi Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- School of Mathematics and Statistics, Shandong University, Weihai 364209, China
| | - Yuelei Zhang
- School of Mathematics and Statistics, Shandong University, Weihai 364209, China
| | - Xiao Chang
- Institute of Statistics and Applied Mathematics, Anhui University of Finance and Economics, Bengbu 233030, China
| | - Xiaoping Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
16
|
Qiu B, Zhang T, Qin X, Ma S, Wang Q. The immune factors have complex causal regulation effects on inflammatory bowel disease. Front Immunol 2024; 14:1322673. [PMID: 38264669 PMCID: PMC10803565 DOI: 10.3389/fimmu.2023.1322673] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/11/2023] [Indexed: 01/25/2024] Open
Abstract
Background Although a correlation between immune cell phenotypes and inflammatory bowel disease (IBD) has been established, a causal relationship remains unestablished. Methods To assess causal associations between immune cell phenotypes and IBD and its subtypes, we employed Mendelian randomization (MR) methods and genome-wide association studies (GWAS) summary statistics. The primary outcomes were determined based on the inverse variance weighting (IVW) results, with the assessment of heterogeneity and pleiotropy conducted through Cochrane's Q-test and MR-Egger. The stability of the MR results was then examined using leave-one-out analysis, and false discovery rate (FDR) correction was applied to evaluate the strength of the causal relationship between exposure and outcome. Furthermore, to identify immunophenotypes strongly associated with IBD, a meta-integration of the effect values of all positive results in both datasets was conducted. Results The analysis of 731 immune cell phenotypes and IBD using MR techniques revealed potential causal associations between 26 phenotypes and IBD. Subsequent meta-integration of the two datasets provided evidence of solid causal associations between 18 immune phenotypes and IBD and its subtypes. Nominal causal associations were also identified in the remaining eight immune phenotypes and IBD and its subtypes. Conclusion Our study confirms causal solid associations between 18 immune phenotypes and IBD, thus guiding future clinical studies.
Collapse
Affiliation(s)
| | | | | | | | - Quan Wang
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
17
|
Funk MA, Leitner J, Gerner MC, Hammerler JM, Salzer B, Lehner M, Battin C, Gumpelmair S, Stiasny K, Grabmeier-Pfistershammer K, Steinberger P. Interrogating ligand-receptor interactions using highly sensitive cellular biosensors. Nat Commun 2023; 14:7804. [PMID: 38016944 PMCID: PMC10684770 DOI: 10.1038/s41467-023-43589-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 11/14/2023] [Indexed: 11/30/2023] Open
Abstract
Interactions of membrane-resident proteins are important targets for therapeutic interventions but most methods to study them are either costly, laborious or fail to reflect the physiologic interaction of membrane resident proteins in trans. Here we describe highly sensitive cellular biosensors as a tool to study receptor-ligand pairs. They consist of fluorescent reporter cells that express chimeric receptors harboring ectodomains of cell surface molecules and intracellular signaling domains. We show that a broad range of molecules can be integrated into this platform and we demonstrate its applicability to highly relevant research areas, including the characterization of immune checkpoints and the probing of cells for the presence of receptors or ligands. The platform is suitable to evaluate the interactions of viral proteins with host receptors and to test for neutralization capability of drugs or biological samples. Our results indicate that cellular biosensors have broad utility as a tool to study protein-interactions.
Collapse
Affiliation(s)
- Maximilian A Funk
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria
| | - Judith Leitner
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria.
| | - Marlene C Gerner
- Division of Biomedical Science, University of Applied Sciences FH Campus Wien, Vienna, Austria
| | - Jasmin M Hammerler
- Division of Biomedical Science, University of Applied Sciences FH Campus Wien, Vienna, Austria
| | - Benjamin Salzer
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- Christian Doppler Laboratory for Next Generation CAR T Cells, Vienna, Austria
| | - Manfred Lehner
- St. Anna Children's Cancer Research Institute, Vienna, Austria
- Christian Doppler Laboratory for Next Generation CAR T Cells, Vienna, Austria
| | - Claire Battin
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria
| | - Simon Gumpelmair
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | - Peter Steinberger
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division for Immune Receptors and T cell activation, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
18
|
Meng Y, Garnish SE, Davies KA, Black KA, Leis AP, Horne CR, Hildebrand JM, Hoblos H, Fitzgibbon C, Young SN, Dite T, Dagley LF, Venkat A, Kannan N, Koide A, Koide S, Glukhova A, Czabotar PE, Murphy JM. Phosphorylation-dependent pseudokinase domain dimerization drives full-length MLKL oligomerization. Nat Commun 2023; 14:6804. [PMID: 37884510 PMCID: PMC10603135 DOI: 10.1038/s41467-023-42255-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/04/2023] [Indexed: 10/28/2023] Open
Abstract
The necroptosis pathway is a lytic, pro-inflammatory mode of cell death that is widely implicated in human disease, including renal, pulmonary, gut and skin inflammatory pathologies. The precise mechanism of the terminal steps in the pathway, where the RIPK3 kinase phosphorylates and triggers a conformation change and oligomerization of the terminal pathway effector, MLKL, are only emerging. Here, we structurally identify RIPK3-mediated phosphorylation of the human MLKL activation loop as a cue for MLKL pseudokinase domain dimerization. MLKL pseudokinase domain dimerization subsequently drives formation of elongated homotetramers. Negative stain electron microscopy and modelling support nucleation of the MLKL tetramer assembly by a central coiled coil formed by the extended, ~80 Å brace helix that connects the pseudokinase and executioner four-helix bundle domains. Mutational data assert MLKL tetramerization as an essential prerequisite step to enable the release and reorganization of four-helix bundle domains for membrane permeabilization and cell death.
Collapse
Affiliation(s)
- Yanxiang Meng
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Sarah E Garnish
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Katherine A Davies
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Katrina A Black
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Andrew P Leis
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Christopher R Horne
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Joanne M Hildebrand
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Hanadi Hoblos
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Cheree Fitzgibbon
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Samuel N Young
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Toby Dite
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Laura F Dagley
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Aarya Venkat
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
| | - Natarajan Kannan
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Akiko Koide
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, 10016, USA
- Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Shohei Koide
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, 10016, USA
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Alisa Glukhova
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Peter E Czabotar
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.
| |
Collapse
|
19
|
Cusick JK, Alcaide J, Shi Y. The RELT Family of Proteins: An Increasing Awareness of Their Importance for Cancer, the Immune System, and Development. Biomedicines 2023; 11:2695. [PMID: 37893069 PMCID: PMC10603948 DOI: 10.3390/biomedicines11102695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
This review highlights Receptor Expressed in Lymphoid Tissues (RELT), a Tumor Necrosis Factor Superfamily member, and its two paralogs, RELL1 and RELL2. Collectively, these three proteins are referred to as RELTfms and have gained much interest in recent years due to their association with cancer and other human diseases. A thorough knowledge of their physiological functions, including the ligand for RELT, is lacking, yet emerging evidence implicates RELTfms in a variety of processes including cytokine signaling and pathways that either promote cell death or survival. T cells from mice lacking RELT exhibit increased responses against tumors and increased inflammatory cytokine production, and multiple lines of evidence indicate that RELT may promote an immunosuppressive environment for tumors. The relationship of individual RELTfms in different cancers is not universal however, as evidence indicates that individual RELTfms may be risk factors in certain cancers yet appear to be protective in other cancers. RELTfms are important for a variety of additional processes related to human health including microbial pathogenesis, inflammation, behavior, reproduction, and development. All three proteins have been strongly conserved in all vertebrates, and this review aims to provide a clearer understanding of the current knowledge regarding these interesting proteins.
Collapse
Affiliation(s)
- John K. Cusick
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
| | - Jessa Alcaide
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
| | - Yihui Shi
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
- California Pacific Medical Center Research Institute, Sutter Bay Hospitals, San Francisco, CA 94107, USA
| |
Collapse
|
20
|
Garnish SE, Martin KR, Kauppi M, Jackson VE, Ambrose R, Eng VV, Chiou S, Meng Y, Frank D, Tovey Crutchfield EC, Patel KM, Jacobsen AV, Atkin-Smith GK, Di Rago L, Doerflinger M, Horne CR, Hall C, Young SN, Cook M, Athanasopoulos V, Vinuesa CG, Lawlor KE, Wicks IP, Ebert G, Ng AP, Slade CA, Pearson JS, Samson AL, Silke J, Murphy JM, Hildebrand JM. A common human MLKL polymorphism confers resistance to negative regulation by phosphorylation. Nat Commun 2023; 14:6046. [PMID: 37770424 PMCID: PMC10539340 DOI: 10.1038/s41467-023-41724-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 09/13/2023] [Indexed: 09/30/2023] Open
Abstract
Across the globe, 2-3% of humans carry the p.Ser132Pro single nucleotide polymorphism in MLKL, the terminal effector protein of the inflammatory form of programmed cell death, necroptosis. Here we show that this substitution confers a gain in necroptotic function in human cells, with more rapid accumulation of activated MLKLS132P in biological membranes and MLKLS132P overriding pharmacological and endogenous inhibition of MLKL. In mouse cells, the equivalent Mlkl S131P mutation confers a gene dosage dependent reduction in sensitivity to TNF-induced necroptosis in both hematopoietic and non-hematopoietic cells, but enhanced sensitivity to IFN-β induced death in non-hematopoietic cells. In vivo, MlklS131P homozygosity reduces the capacity to clear Salmonella from major organs and retards recovery of hematopoietic stem cells. Thus, by dysregulating necroptosis, the S131P substitution impairs the return to homeostasis after systemic challenge. Present day carriers of the MLKL S132P polymorphism may be the key to understanding how MLKL and necroptosis modulate the progression of complex polygenic human disease.
Collapse
Affiliation(s)
- Sarah E Garnish
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Katherine R Martin
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Maria Kauppi
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Victoria E Jackson
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Rebecca Ambrose
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Vik Ven Eng
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Shene Chiou
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Yanxiang Meng
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Daniel Frank
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Emma C Tovey Crutchfield
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, Parkville, VIC, Australia
| | - Komal M Patel
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Annette V Jacobsen
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Georgia K Atkin-Smith
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Ladina Di Rago
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Marcel Doerflinger
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Christopher R Horne
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Cathrine Hall
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Samuel N Young
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Matthew Cook
- Centre for Personalised Immunology and Canberra Clinical Genomics, Australian National University, Canberra, ACT, Australia
- Cambridge Institute for Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
| | - Vicki Athanasopoulos
- Department of Immunology and Infection, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Carola G Vinuesa
- Centre for Personalised Immunology and Canberra Clinical Genomics, Australian National University, Canberra, ACT, Australia
- Department of Immunology and Infection, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- The Francis Crick Institute, London, UK
- University College London, London, UK
- China Australia Centre for Personalized Immunology (CACPI), Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Kate E Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Ian P Wicks
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Gregor Ebert
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Ashley P Ng
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
- Clinical Haematology Department, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Charlotte A Slade
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
- Department of Clinical Immunology & Allergy, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Jaclyn S Pearson
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - André L Samson
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - John Silke
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - James M Murphy
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Joanne M Hildebrand
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.
- University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia.
| |
Collapse
|
21
|
Wenker SD, Farias MI, Gradaschi V, Garcia C, Beauquis J, Leal MC, Ferrari C, Zeng X, Pitossi FJ. Microglia-secreted TNF-α affects differentiation efficiency and viability of pluripotent stem cell-derived human dopaminergic precursors. PLoS One 2023; 18:e0263021. [PMID: 37751438 PMCID: PMC10521980 DOI: 10.1371/journal.pone.0263021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 08/19/2023] [Indexed: 09/28/2023] Open
Abstract
Disease is a neurodegenerative disorder characterised by the progressive loss of dopaminergic cells of the substantia nigra pars compacta. Even though successful transplantation of dopamine-producing cells into the striatum exhibits favourable effects in animal models and clinical trials; transplanted cell survival is low. Since every transplant elicits an inflammatory response which can affect cell survival and differentiation, we aimed to study in vivo and in vitro the impact of the pro-inflammatory environment on human dopaminergic precursors. We first observed that transplanted human dopaminergic precursors into the striatum of immunosuppressed rats elicited an early and sustained activation of astroglial and microglial cells after 15 days' post-transplant. This long-lasting response was associated with Tumour necrosis factor alpha expression in microglial cells. In vitro, conditioned media from activated BV2 microglial cells increased cell death, decreased Tyrosine hydroxylase-positive cells and induced morphological alterations on human neural stem cells-derived dopaminergic precursors at two differentiation stages: 19 days and 28 days. Those effects were ameliorated by inhibition of Tumour necrosis factor alpha, a cytokine which was previously detected in vivo and in conditioned media from activated BV-2 cells. Our results suggest that a pro-inflammatory environment is sustained after transplantation under immunosuppression, providing a window of opportunity to modify this response to increase transplant survival and differentiation. In addition, our data show that the microglia-derived pro-inflammatory microenvironment has a negative impact on survival and differentiation of dopaminergic precursors. Finally, Tumour necrosis factor alpha plays a key role in these effects, suggesting that this cytokine could be an interesting target to increase the efficacy of human dopaminergic precursors transplantation in Parkinson's Disease.
Collapse
Affiliation(s)
| | | | | | - Corina Garcia
- Fundación Instituto Leloir—IIBBA-CONICET, Buenos Aires, Argentina
| | - Juan Beauquis
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Carina Ferrari
- Fundación Instituto Leloir—IIBBA-CONICET, Buenos Aires, Argentina
| | | | | |
Collapse
|
22
|
Wang X, Wang L, Liu W, Liu X, Jia X, Feng X, Li F, Zhu R, Yu J, Zhang H, Wu H, Wu J, Wang C, Yu B, Yu X. Dose-related immunomodulatory effects of recombinant TRAIL in the tumor immune microenvironment. J Exp Clin Cancer Res 2023; 42:216. [PMID: 37605148 PMCID: PMC10464183 DOI: 10.1186/s13046-023-02795-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/10/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND In addition to specifically inducing tumor cell apoptosis, recombinant tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has also been reported to influence the cancer immune microenvironment; however, its underlying effects and mechanisms remain unclear. Investigating the immunomodulatory effects and mechanisms of recombinant TRAIL in the tumor microenvironment (TME) may provide an important perspective and facilitate the exploration of novel TRAIL strategies for tumor therapy. METHODS Immunocompetent mice with different tumors were treated with three doses of recombinant TRAIL, and then the tumors were collected for immunological detection and mechanistic investigation. Methodological approaches include flow cytometry analysis and single-cell sequencing. RESULTS In an immunocompetent mouse model, recombinant soluble mouse TRAIL (smTRAIL) had dose-related immunomodulatory effects. The optimal dose of smTRAIL (2 mg/kg) activated innate immune cells and CD8+ T cells, whereas higher doses of smTRAIL (8 mg/kg) promoted the formation of a tumor-promoting immune microenvironment to counteract the apoptotic effects on tumor cells. The higher doses of smTRAIL treatment promoted M2-like macrophage recruitment and polarization and increased the production of protumor inflammatory cytokines, such as IL-10, which deepened the suppression of natural killer (NK) cells and CD8+ T cells in the tumor microenvironment. By constructing an HU-HSC-NPG.GM3 humanized immune system mouse model, we further verified the immunomodulatory effects induced by recombinant soluble human TRAIL (shTRAIL) and found that combinational administration of shTRAIL and trabectedin, a macrophage-targeting drug, could remodel the tumor immune microenvironment, further enhance antitumor immunity, and strikingly improve antitumor effects. CONCLUSION Our results highlight the immunomodulatory role of recombinant TRAIL and suggest promising therapeutic strategies for clinical application.
Collapse
Affiliation(s)
- Xupu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Lizheng Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - Wenmo Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Xinyao Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Xinyuan Jia
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Xinyao Feng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Fangshen Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Rui Zhu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Jiahao Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Haihong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Hui Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Jiaxin Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Chu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China.
| |
Collapse
|
23
|
Wenthe J, Eriksson E, Hellström AC, Moreno R, Ullenhag G, Alemany R, Lövgren T, Loskog A. Immunostimulatory gene therapy targeting CD40, 4-1BB and IL-2R activates DCs and stimulates antigen-specific T-cell and NK-cell responses in melanoma models. J Transl Med 2023; 21:506. [PMID: 37501121 PMCID: PMC10373363 DOI: 10.1186/s12967-023-04374-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND The activation of dendritic cells (DCs) is pivotal for generating antigen-specific T-cell responses to eradicate tumor cells. Hence, immunotherapies targeting this interplay are especially intriguing. Moreover, it is of interest to modulate the tumor microenvironment (TME), as this harsh milieu often impairs adaptive immune responses. Oncolytic viral therapy presents an opportunity to overcome the immunosuppression in tumors by destroying tumor cells and thereby releasing antigens and immunostimulatory factors. These effects can be further amplified by the introduction of transgenes expressed by the virus. METHODS Lokon oncolytic adenoviruses (LOAd) belong to a platform of chimeric serotype Ad5/35 viruses that have their replication restricted to tumor cells, but the expression of transgenes is permitted in all infected cells. LOAd732 is a novel oncolytic adenovirus that expresses three essential immunostimulatory transgenes: trimerized membrane-bound CD40L, 4-1BBL and IL-2. Transgene expression was determined with flow cytometry and ELISA and the oncolytic function was evaluated with viability assays and xenograft models. The activation profiles of DCs were investigated in co-cultures with tumor cells or in an autologous antigen-specific T cell model by flow cytometry and multiplex proteomic analysis. Statistical differences were analyzed with Kruskal-Wallis test followed by Dunn's multiple comparison test. RESULTS All three transgenes were expressed in infected melanoma cells and DCs and transgene expression did not impair the oncolytic activity in tumor cells. DCs were matured post LOAd732 infection and expressed a multitude of co-stimulatory molecules and pro-inflammatory cytokines crucial for T-cell responses. Furthermore, these DCs were capable of expanding and stimulating antigen-specific T cells in addition to natural killer (NK) cells. Strikingly, the addition of immunosuppressive cytokines TGF-β1 and IL-10 did not affect the ability of LOAd732-matured DCs to expand antigen-specific T cells and these cells retained an enhanced activation profile. CONCLUSIONS LOAd732 is a novel immunostimulatory gene therapy based on an oncolytic adenovirus that expresses three transgenes, which are essential for mediating an anti-tumor immune response by activating DCs and stimulating T and NK cells even under imunosuppressive conditions commonly present in the TME. These qualities make LOAd732 an appealing new immunotherapy approach.
Collapse
Affiliation(s)
- Jessica Wenthe
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 751 85, Uppsala, Sweden.
- Lokon Pharma AB, Uppsala, Sweden.
| | - Emma Eriksson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 751 85, Uppsala, Sweden
- Lokon Pharma AB, Uppsala, Sweden
| | - Ann-Charlotte Hellström
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 751 85, Uppsala, Sweden
| | - Rafael Moreno
- IDIBELL-Institute Català d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Gustav Ullenhag
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 751 85, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Ramon Alemany
- IDIBELL-Institute Català d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Tanja Lövgren
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 751 85, Uppsala, Sweden
| | - Angelica Loskog
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsväg 20, 751 85, Uppsala, Sweden
- Lokon Pharma AB, Uppsala, Sweden
| |
Collapse
|
24
|
Liu W, Gao L, Hou X, Feng S, Yan H, Pan H, Zhang S, Yang X, Jiang J, Ye F, Zhao Q, Wei L, Han Z. TWEAK Signaling-Induced ID1 Expression Drives Malignant Transformation of Hepatic Progenitor Cells During Hepatocarcinogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300350. [PMID: 37085918 PMCID: PMC10288241 DOI: 10.1002/advs.202300350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/14/2023] [Indexed: 05/03/2023]
Abstract
The malignant transformation of hepatic progenitor cells (HPCs) in the inflammatory microenvironment is the root cause of hepatocarcinogenesis. However, the potential molecular mechanisms are still elusive. The HPCs subgroup is identified by single-cell RNA (scRNA) sequencing and the phenotype of HPCs is investigated in the primary HCC model. Bulk RNA sequencing (RNA-seq) and proteomic analyses are also performed on HPC-derived organoids. It is found that tumors are formed from HPCs in peritumor tissue at the 16th week in a HCC model. Furthermore, it is confirmed that the macrophage-derived TWEAK/Fn14 promoted the expression of inhibitor of differentiation-1 (ID1) in HPCs via NF-κB signaling and a high level of ID1 induced aberrant differentiation of HPCs. Mechanistically, ID1 suppressed differentiation and promoted proliferation in HPCs through the inhibition of HNF4α and Rap1GAP transcriptions. Finally, scRNA sequencing of HCC patients and investigation of clinical specimens also verified that the expression of ID1 is correlated with aberrant differentiation of HPCs into cancer stem cells, patients with high levels of ID1 in HPCs showed a poorer prognosis. This study provides important intervention targets and a theoretical basis for the clinical diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Wenting Liu
- Tumor Immunology and Gene Therapy CenterThird Affiliated Hospital of Naval Medical UniversityShanghai200438P. R. China
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer of Ministry of EducationEastern Hepatobiliary Surgery Hospital/National Center for Liver CancerNaval Medical UniversityShanghai200438P. R. China
| | - Lu Gao
- Tumor Immunology and Gene Therapy CenterThird Affiliated Hospital of Naval Medical UniversityShanghai200438P. R. China
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer of Ministry of EducationEastern Hepatobiliary Surgery Hospital/National Center for Liver CancerNaval Medical UniversityShanghai200438P. R. China
| | - Xiaojuan Hou
- Tumor Immunology and Gene Therapy CenterThird Affiliated Hospital of Naval Medical UniversityShanghai200438P. R. China
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer of Ministry of EducationEastern Hepatobiliary Surgery Hospital/National Center for Liver CancerNaval Medical UniversityShanghai200438P. R. China
| | - Shiyao Feng
- Department of UrologySecond Affiliated HospitalAnhui Medical UniversityHefei230601P. R. China
| | - Haixin Yan
- Department of UrologySecond Affiliated HospitalAnhui Medical UniversityHefei230601P. R. China
| | - Hongyu Pan
- Department of Hepatic SurgeryThird Affiliated Hospital of Naval Medical UniversityShanghai200438P. R. China
| | - Shichao Zhang
- Department of Hepatic SurgeryThird Affiliated Hospital of Naval Medical UniversityShanghai200438P. R. China
| | - Xue Yang
- Tumor Immunology and Gene Therapy CenterThird Affiliated Hospital of Naval Medical UniversityShanghai200438P. R. China
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer of Ministry of EducationEastern Hepatobiliary Surgery Hospital/National Center for Liver CancerNaval Medical UniversityShanghai200438P. R. China
| | - Jinghua Jiang
- Tumor Immunology and Gene Therapy CenterThird Affiliated Hospital of Naval Medical UniversityShanghai200438P. R. China
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer of Ministry of EducationEastern Hepatobiliary Surgery Hospital/National Center for Liver CancerNaval Medical UniversityShanghai200438P. R. China
| | - Fei Ye
- Tumor Immunology and Gene Therapy CenterThird Affiliated Hospital of Naval Medical UniversityShanghai200438P. R. China
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer of Ministry of EducationEastern Hepatobiliary Surgery Hospital/National Center for Liver CancerNaval Medical UniversityShanghai200438P. R. China
| | - Qiudong Zhao
- Tumor Immunology and Gene Therapy CenterThird Affiliated Hospital of Naval Medical UniversityShanghai200438P. R. China
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer of Ministry of EducationEastern Hepatobiliary Surgery Hospital/National Center for Liver CancerNaval Medical UniversityShanghai200438P. R. China
| | - Lixin Wei
- Tumor Immunology and Gene Therapy CenterThird Affiliated Hospital of Naval Medical UniversityShanghai200438P. R. China
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer of Ministry of EducationEastern Hepatobiliary Surgery Hospital/National Center for Liver CancerNaval Medical UniversityShanghai200438P. R. China
| | - Zhipeng Han
- Tumor Immunology and Gene Therapy CenterThird Affiliated Hospital of Naval Medical UniversityShanghai200438P. R. China
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer of Ministry of EducationEastern Hepatobiliary Surgery Hospital/National Center for Liver CancerNaval Medical UniversityShanghai200438P. R. China
| |
Collapse
|
25
|
Frankish J, Mukherjee D, Romano E, Billian-Frey K, Schröder M, Heinonen K, Merz C, Redondo Müller M, Gieffers C, Hill O, Thiemann M, Honeychurch J, Illidge T, Sykora J. The CD40 agonist HERA-CD40L results in enhanced activation of antigen presenting cells, promoting an anti-tumor effect alone and in combination with radiotherapy. Front Immunol 2023; 14:1160116. [PMID: 37304285 PMCID: PMC10251205 DOI: 10.3389/fimmu.2023.1160116] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction The ability to modulate and enhance the anti-tumor immune responses is critical in developing novel therapies in cancer. The Tumor Necrosis Factor (TNF) Receptor Super Family (TNFRSF) are potentially excellent targets for modulation which result in specific anti-tumor immune responses. CD40 is a member of the TNFRSF and several clinical therapies are under development. CD40 signaling plays a pivotal role in regulating the immune system from B cell responses to myeloid cell driven activation of T cells. The CD40 signaling axis is well characterized and here we compare next generation HERA-Ligands to conventional monoclonal antibody based immune modulation for the treatment of cancer. Methods & results HERA-CD40L is a novel molecule that targets CD40 mediated signal transduction and demonstrates a clear mode of action in generating an activated receptor complex via recruitment of TRAFs, cIAP1, and HOIP, leading to TRAF2 phosphorylation and ultimately resulting in the enhanced activation of key inflammatory/survival pathway and transcription factors such asNFkB, AKT, p38, ERK1/2, JNK, and STAT1 in dendritic cells. Furthermore, HERA-CD40L demonstrated a strong modulation of the tumor microenvironment (TME) via the increase in intratumoral CD8+ T cells and the functional switch from pro-tumor macrophages (TAMs) to anti-tumor macrophages that together results in a significant reduction of tumor growth in a CT26 mouse model. Furthermore, radiotherapy which may have an immunosuppressive modulation of the TME, was shown to have an immunostimulatory effect in combination with HERA-CD40L. Radiotherapy in combination with HERA-CD40L treatment resulted in an increase in detected intratumoral CD4+/8+ T cells compared to RT alone and, additionally, the repolarization of TAMs was also observed, resulting in an inhibition of tumor growth in a TRAMP-C1 mouse model. Discussion Taken together, HERA-CD40L resulted in activating signal transduction mechanisms in dendritic cells, resulting in an increase in intratumoral T cells and manipulation of the TME to be pro-inflammatory, repolarizing M2 macrophages to M1, enhancing tumor control.
Collapse
Affiliation(s)
| | - Debayan Mukherjee
- Targeted Therapy Group, Division of Cancer Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Erminia Romano
- Targeted Therapy Group, Division of Cancer Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | | - Jamie Honeychurch
- Targeted Therapy Group, Division of Cancer Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Tim Illidge
- Targeted Therapy Group, Division of Cancer Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | | |
Collapse
|
26
|
Dhusia K, Su Z, Wu Y. Computational analyses of the interactome between TNF and TNFR superfamilies. Comput Biol Chem 2023; 103:107823. [PMID: 36682326 DOI: 10.1016/j.compbiolchem.2023.107823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/05/2023] [Accepted: 01/18/2023] [Indexed: 01/20/2023]
Abstract
Proteins in the tumor necrosis factor (TNF) superfamily (TNFSF) regulate diverse cellular processes by interacting with their receptors in the TNF receptor (TNFR) superfamily (TNFRSF). Ligands and receptors in these two superfamilies form a complicated network of interactions, in which the same ligand can bind to different receptors and the same receptor can be shared by different ligands. In order to study these interactions on a systematic level, a TNFSF-TNFRSF interactome was constructed in this study by searching the database which consists of both experimentally measured and computationally predicted protein-protein interactions (PPIs). The interactome contains a total number of 194 interactions between 18 TNFSF ligands and 29 TNFRSF receptors in human. We modeled the structure for each ligand-receptor interaction in the network. Their binding affinities were further computationally estimated based on modeled structures. Our computational outputs, which are all publicly accessible, serve as a valuable addition to the currently limited experimental resources to study TNF-mediated cell signaling.
Collapse
Affiliation(s)
- Kalyani Dhusia
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, the United States of America
| | - Zhaoqian Su
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, the United States of America
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, the United States of America.
| |
Collapse
|
27
|
Tovey Crutchfield EC, Garnish SE, Day J, Anderton H, Chiou S, Hempel A, Hall C, Patel KM, Gangatirkar P, Martin KR, Li Wai Suen CSN, Garnham AL, Kueh AJ, Wicks IP, Silke J, Nachbur U, Samson AL, Murphy JM, Hildebrand JM. MLKL deficiency protects against low-grade, sterile inflammation in aged mice. Cell Death Differ 2023; 30:1059-1071. [PMID: 36755069 PMCID: PMC10070424 DOI: 10.1038/s41418-023-01121-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/16/2023] [Accepted: 01/23/2023] [Indexed: 02/10/2023] Open
Abstract
MLKL and RIPK3 are the core signaling proteins of the inflammatory cell death pathway, necroptosis, which is a known mediator and modifier of human disease. Necroptosis has been implicated in the progression of disease in almost every physiological system and recent reports suggest a role for necroptosis in aging. Here, we present the first comprehensive analysis of age-related histopathological and immunological phenotypes in a cohort of Mlkl-/- and Ripk3-/- mice on a congenic C57BL/6 J genetic background. We show that genetic deletion of Mlkl in female mice interrupts immune system aging, specifically delaying the age-related reduction of circulating lymphocytes. -Seventeen-month-old Mlkl-/- female mice were also protected against age-related chronic sterile inflammation in connective tissue and skeletal muscle relative to wild-type littermate controls, exhibiting a reduced number of immune cell infiltrates in these sites and fewer regenerating myocytes. These observations implicate MLKL in age-related sterile inflammation, suggesting a possible application for long-term anti-necroptotic therapy in humans.
Collapse
Affiliation(s)
- Emma C Tovey Crutchfield
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.,The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia.,The University of Melbourne, Faculty of Medicine, Dentistry and Health Sciences, Parkville, VIC, Australia
| | - Sarah E Garnish
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.,The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Jessica Day
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.,The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia.,Royal Melbourne Hospital, Rheumatology Unit, Parkville, VIC, Australia
| | - Holly Anderton
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.,The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Shene Chiou
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.,The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Anne Hempel
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Cathrine Hall
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | - Komal M Patel
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia
| | | | - Katherine R Martin
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.,The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | | | | | - Andrew J Kueh
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.,The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Ian P Wicks
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.,The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia.,Royal Melbourne Hospital, Rheumatology Unit, Parkville, VIC, Australia
| | - John Silke
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.,The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Ueli Nachbur
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.,The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - Andre L Samson
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.,The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia
| | - James M Murphy
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia. .,The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia.
| | - Joanne M Hildebrand
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia. .,The University of Melbourne, Department of Medical Biology, Parkville, VIC, Australia.
| |
Collapse
|
28
|
Martin KR, Day JA, Hansen JA, D'Silva DB, Wong HL, Garnham A, Sandow JJ, Nijagal B, Wilson N, Wicks IP. CD98 defines a metabolically flexible, proinflammatory subset of low-density neutrophils in systemic lupus erythematosus. Clin Transl Med 2023; 13:e1150. [PMID: 36653319 PMCID: PMC9849148 DOI: 10.1002/ctm2.1150] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Low-density neutrophils (LDN) are a distinct subset of neutrophils rarely detected in healthy people but appear in the blood of patients with autoimmune diseases, including systemic lupus erythematosus (SLE), and are mobilised in response to granulocyte colony-stimulating factor (G-CSF). The aim of this study was to identify novel mechanisms responsible for the pathogenic capacity of LDN in SLE. METHODS Neutrophils were isolated from donors treated with G-CSF, and whole-cell proteomic analysis was performed on LDN and normal-density neutrophils. RESULTS CD98 is significantly upregulated in LDN from G-CSF donors and defines a subset of LDN within the blood of SLE patients. CD98 is a transmembrane protein that dimerises with L-type amino acid transporters. We show that CD98 is responsible for the increased bioenergetic capacity of LDN. CD98 on LDN mediates the uptake of essential amino acids that are used by mitochondria to produce adenosine triphosphate, especially in the absence of glucose. Inhibition of CD98 reduces the metabolic flexibility of this population, which may limit their pathogenic capacity. CD98+ LDN produce more proinflammatory cytokines and chemokines than their normal density counterparts and are resistant to apoptosis, which may also contribute to tissue inflammation and end organ damage in SLE. CONCLUSIONS CD98 provides a phenotypic marker for LDN that facilitates identification of this population without density-gradient separation and represents a novel therapeutic target to limit its pathogenic capacity.
Collapse
Affiliation(s)
- Katherine R. Martin
- Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoriaAustralia
| | - Jessica A. Day
- Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoriaAustralia
- Department of RheumatologyRoyal Melbourne HospitalParkvilleVictoriaAustralia
| | - Jacinta A. Hansen
- Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
| | - Damian B. D'Silva
- Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
| | - Huon L. Wong
- Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
| | - Alexandra Garnham
- Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoriaAustralia
| | - Jarrod J. Sandow
- Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoriaAustralia
| | - Brunda Nijagal
- Metabolomics AustraliaBio21 Institute of Molecular Science and BiotechnologyUniversity of MelbourneParkvilleVictoriaAustralia
| | | | - Ian P. Wicks
- Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoriaAustralia
- Department of RheumatologyRoyal Melbourne HospitalParkvilleVictoriaAustralia
| |
Collapse
|
29
|
Eslami M, Willen D, Papasouliotis O, Schuepbach-Mallpell S, Willen L, Donzé O, Yalkinoglu Ö, Schneider P. Kinetics of free and ligand-bound atacicept in human serum. Front Immunol 2022; 13:1035556. [PMID: 36532058 PMCID: PMC9756848 DOI: 10.3389/fimmu.2022.1035556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
BAFF (B cell activation factor of the TNF family/B lymphocyte stimulator, BLyS) and APRIL (a proliferation-inducing ligand) are targeted by atacicept, a decoy receptor consisting of the extracellular domain of TACI (transmembrane activator and calcium-modulator and cyclophilin (CAML) interactor) fused to the Fc portion of human IgG1. The purpose of the study was to characterize free and ligand-bound atacicept in humans. Total and active atacicept in serum of healthy volunteers receiving a single dose of subcutaneous atacicept or in patients treated weekly for one year were measured by ELISA, Western blot, or cell-based assays. Pharmacokinetics of free and bound atacicept were predicted based on total atacicept ELISA results. Persistence of complexes of purified atacicept bound to recombinant ligands was also monitored in mice. Results show that unbound or active atacicept in human serum exceeded 0.1 µg/ml for one week post administration, or throughout a 1-year treatment with weekly administrations. After a single administration of atacicept, endogenous BAFF bound to atacicept was detected after 8 h then increased about 100-fold within 2 to 4 weeks. Endogenous heteromers of BAFF and APRIL bound to atacicept also accumulated, but atacicept-APRIL complexes were not detected. In mice receiving intravenous injections of purified complexes pre-formed in vitro, atacicept-BAFF persisted longer (more than a week) than atacicept-APRIL (less than a day). Thus, only biologically inactive BAFF and BAFF-APRIL heteromers accumulate on atacicept in vivo. The measure of active atacicept provides further support for the once-weekly dosing regimen implemented in the clinical development of atacicept.
Collapse
Affiliation(s)
- Mahya Eslami
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Daniela Willen
- Clinical Pharmacology, Translational Medicine, Merck Healthcare KGaA, Darmstadt, Germany
| | - Orestis Papasouliotis
- Translational Medicine, Merck Institute for Pharmacometrics (an affiliate of Merck KGaA), Lausanne, Switzerland
| | | | - Laure Willen
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | | | - Özkan Yalkinoglu
- Clinical Pharmacology, Translational Medicine, Merck Healthcare KGaA, Darmstadt, Germany
| | - Pascal Schneider
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland,*Correspondence: Pascal Schneider,
| |
Collapse
|
30
|
Sato A, Azuma M, Nagai H, Imai W, Kawaguchi K, Morita M, Okuyama Y, Ishii N, So T. OX40 Ligand-Mannose-Binding Lectin Fusion Protein Induces Potent OX40 Cosignaling in CD4 + T Cells. Biol Pharm Bull 2022; 45:1798-1804. [PMID: 36450532 DOI: 10.1248/bpb.b22-00493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
OX40, a member of the tumor necrosis factor (TNF) receptor superfamily, is induced on activated T cells. Membrane-bound OX40 ligand (OX40L) expressed by activated antigen-presenting cells induces OX40 signaling, which promotes T cell immunity. OX40 agonism would be a potential target for immunotherapy, however, it remains unclear how the activity of OX40 can be successfully controlled by a designer OX40L protein. We prepared a soluble OX40L protein possessing a PA-peptide tag and a collagenous trimerization domain from mannose-binding lectin (MBL), and tested whether PA-MBL-OX40L fusion protein worked as an agonist for OX40. We found that the majority of recombinant PA-MBL-OX40L protein purified from culture supernatants displayed a trimer structure and bound to cell surface OX40 or OX40-Fc fusion protein in a dose-dependent manner. Upon stimulation of CD4+ T cells with TCR/CD3 without CD28, PA-MBL-OX40L displayed significantly increased proliferative and cytokine responses when compared with a benchmark agonistic monoclonal antibody for OX40. Both soluble and immobilized forms of PA-MBL-OX40L induced potent OX40 signaling in CD4+ T cells. Mice administered with PA-MBL-OX40L displayed significantly augmented T cell-mediated delayed-type hypersensitivity responses. Our results suggest that activity of OX40L could be engineered to elicit better T cell responses by rational design of its assembly and architecture.
Collapse
Affiliation(s)
- Ayaka Sato
- Laboratory of Molecular Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Mitsuki Azuma
- Laboratory of Molecular Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Hodaka Nagai
- Laboratory of Molecular Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Wakana Imai
- Laboratory of Molecular Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Kosuke Kawaguchi
- Laboratory of Molecular Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Masashi Morita
- Laboratory of Molecular Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Yuko Okuyama
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Tohoku University
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Tohoku University
| | - Takanori So
- Laboratory of Molecular Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama.,Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Tohoku University
| |
Collapse
|
31
|
Zwolak A, Chan SR, Harvilla P, Mahady S, Armstrong AA, Luistro L, Tamot N, Yamada D, Derebe M, Pomerantz S, Chiu M, Ganesan R, Chowdhury P. A stable, engineered TL1A ligand co-stimulates T cells via specific binding to DR3. Sci Rep 2022; 12:20538. [PMID: 36446890 PMCID: PMC9709071 DOI: 10.1038/s41598-022-24984-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022] Open
Abstract
TL1A (TNFSF15) is a TNF superfamily ligand which can bind the TNFRSF member death receptor 3 (DR3) on T cells and the soluble decoy receptor DcR3. Engagement of DR3 on CD4+ or CD8+ effector T cells by TL1A induces downstream signaling, leading to proliferation and an increase in secretion of inflammatory cytokines. We designed a stable recombinant TL1A molecule that (1) displays high monodispersity and stability, (2) displays the ability to activate T cells in vitro and in vivo, and (3) lacks binding to DcR3 while retaining functional activity via DR3. Together these results suggest the TL1A ligand can be amenable to therapeutic development on its own or paired with a tumor-targeting moiety.
Collapse
Affiliation(s)
- Adam Zwolak
- grid.497530.c0000 0004 0389 4927Biologics Discovery, Janssen Research & Development, LLC, Spring House, PA 19477 USA
| | - Szeman Ruby Chan
- grid.497530.c0000 0004 0389 4927Oncology Discovery, Janssen Research & Development, LLC, Spring House, PA 19477 USA
| | - Paul Harvilla
- grid.497530.c0000 0004 0389 4927Biologics Discovery, Janssen Research & Development, LLC, Spring House, PA 19477 USA
| | - Sally Mahady
- grid.497530.c0000 0004 0389 4927Oncology Discovery, Janssen Research & Development, LLC, Spring House, PA 19477 USA
| | - Anthony A. Armstrong
- grid.497530.c0000 0004 0389 4927Biologics Discovery, Janssen Research & Development, LLC, Spring House, PA 19477 USA
| | - Leopoldo Luistro
- grid.497530.c0000 0004 0389 4927Oncology Discovery, Janssen Research & Development, LLC, Spring House, PA 19477 USA
| | - Ninkka Tamot
- grid.497530.c0000 0004 0389 4927Biologics Discovery, Janssen Research & Development, LLC, Spring House, PA 19477 USA
| | - Douglas Yamada
- grid.497530.c0000 0004 0389 4927Oncology Discovery, Janssen Research & Development, LLC, Spring House, PA 19477 USA
| | - Mehabaw Derebe
- grid.417993.10000 0001 2260 0793Merck Research Laboratories, Discovery Biologics, Protein Sciences, South San Francisco, CA USA
| | - Steven Pomerantz
- grid.497530.c0000 0004 0389 4927Biologics Discovery, Janssen Research & Development, LLC, Spring House, PA 19477 USA
| | - Mark Chiu
- Tavotek Biotherapeutics, Spring House, PA USA
| | - Rajkumar Ganesan
- grid.417886.40000 0001 0657 5612Immunotherapeutics, Amgen, South San Francisco, CA USA
| | - Partha Chowdhury
- grid.497530.c0000 0004 0389 4927Cell Engineering and Early Development, Janssen Research & Development, Spring House, PA USA
| |
Collapse
|
32
|
D Lempicki M, Paul S, Serbulea V, Upchurch CM, Sahu S, Gray JA, Ailawadi G, Garcia BL, McNamara CA, Leitinger N, Meher AK. BAFF antagonism via the BAFF receptor 3 binding site attenuates BAFF 60-mer-induced classical NF-κB signaling and metabolic reprogramming of B cells. Cell Immunol 2022; 381:104603. [PMID: 36182705 PMCID: PMC10691782 DOI: 10.1016/j.cellimm.2022.104603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/03/2022]
Abstract
Human recombinant B cell activating factor (BAFF) is secreted as 3-mers, which can associate to form 60-mers in culture supernatants. However, the presence of BAFF multimers in humans is still debated and it is incompletely understood how BAFF multimers activate the B cells. Here, we demonstrate that BAFF can exist as 60-mers or higher order multimers in human plasma. In vitro, BAFF 60-mer strongly induced the transcriptome of B cells which was partly attenuated by antagonism using a soluble fragment of BAFF receptor 3. Furthermore, compared to BAFF 3-mer, BAFF 60-mer strongly induced a transient classical and prolonged alternate NF-κB signaling, glucose oxidation by both aerobic glycolysis and oxidative phosphorylation, and succinate utilization by mitochondria. BAFF antagonism selectively attenuated classical NF-κB signaling and glucose oxidation. Altogether, our results suggest critical roles of BAFF 60-mer and its BAFF receptor 3 binding site in hyperactivation of B cells.
Collapse
Affiliation(s)
- Melissa D Lempicki
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, United States
| | - Saikat Paul
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, United States
| | - Vlad Serbulea
- Department of Pharmacology, University of Virginia, VA 22908, United States
| | - Clint M Upchurch
- Department of Pharmacology, University of Virginia, VA 22908, United States
| | - Srabani Sahu
- Department of Pharmacology, University of Virginia, VA 22908, United States
| | - Jake A Gray
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, United States
| | - Gorav Ailawadi
- Department of Surgery, University of Virginia, VA 22908, United States
| | - Brandon L Garcia
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, United States
| | - Coleen A McNamara
- Robert M. Berne Cardiovascular Research Center, University of Virginia, VA 22908, United States
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia, VA 22908, United States
| | - Akshaya K Meher
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, United States; Department of Pharmacology, University of Virginia, VA 22908, United States.
| |
Collapse
|
33
|
Sethi A, Horne CR, Fitzgibbon C, Wilde K, Davies KA, Garnish SE, Jacobsen AV, Samson AL, Hildebrand JM, Wardak A, Czabotar PE, Petrie EJ, Gooley PR, Murphy JM. Membrane permeabilization is mediated by distinct epitopes in mouse and human orthologs of the necroptosis effector, MLKL. Cell Death Differ 2022; 29:1804-1815. [PMID: 35264780 PMCID: PMC9433430 DOI: 10.1038/s41418-022-00965-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 02/21/2022] [Accepted: 02/21/2022] [Indexed: 12/18/2022] Open
Abstract
Necroptosis is a lytic programmed cell death pathway with origins in innate immunity that is frequently dysregulated in inflammatory diseases. The terminal effector of the pathway, MLKL, is licensed to kill following phosphorylation of its pseudokinase domain by the upstream regulator, RIPK3 kinase. Phosphorylation provokes the unleashing of MLKL's N-terminal four-helix bundle (4HB or HeLo) domain, which binds and permeabilizes the plasma membrane to cause cell death. The precise mechanism by which the 4HB domain permeabilizes membranes, and how the mechanism differs between species, remains unclear. Here, we identify the membrane binding epitope of mouse MLKL using NMR spectroscopy. Using liposome permeabilization and cell death assays, we validate K69 in the α3 helix, W108 in the α4 helix, and R137/Q138 in the first brace helix as crucial residues for necroptotic signaling. This epitope differs from the phospholipid binding site reported for human MLKL, which comprises basic residues primarily located in the α1 and α2 helices. In further contrast to human and plant MLKL orthologs, in which the α3-α4 loop forms a helix, this loop is unstructured in mouse MLKL in solution. Together, these findings illustrate the versatility of the 4HB domain fold, whose lytic function can be mediated by distinct epitopes in different orthologs.
Collapse
Affiliation(s)
- Ashish Sethi
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Christopher R Horne
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Cheree Fitzgibbon
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Karyn Wilde
- National Deuteration Facility, Australian Nuclear Science and Technology Organization, Lucas Heights, NSW, 2234, Australia
| | - Katherine A Davies
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Sarah E Garnish
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Annette V Jacobsen
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - André L Samson
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Joanne M Hildebrand
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Ahmad Wardak
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Peter E Czabotar
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Emma J Petrie
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Paul R Gooley
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
| |
Collapse
|
34
|
Bolkun L, Tynecka M, Wasiluk T, Piszcz J, Starosz A, Grubczak K, Moniuszko M, Eljaszewicz A. A Proliferation-Inducing Ligand and B-Cell Activating Factor Are Upregulated in Patients with Essential Thrombocythemia. J Clin Med 2022; 11:jcm11164663. [PMID: 36012902 PMCID: PMC9409834 DOI: 10.3390/jcm11164663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 12/20/2022] Open
Abstract
A proliferation-inducing ligand (APRIL) and B-cell activating factor (BAFF) are cytokines belonging to the tumor necrosis factor family which play an essential role in B-cell maturation, differentiation, and survival. Recent evidence indicates their importance in hematological disorders; however, their function in essential thrombocytosis (ET) pathogenesis remains elusive. Therefore, we aimed to analyze the role of APRIL and BAFF in megakaryocytopoiesis in ET patients. We observed elevated levels of APRIL and BAFF in the plasma of ET patients compared with healthy controls, while no differences were found among patients with different JAK2(V617F) statuses. In addition, APRIL levels were positively associated with the number of platelets and WBC count. In the bone marrow, APRIL but not BAFF levels were higher in ET patients with the JAK2(V617F) mutation; however, JAK2(V617F)-negative patients showed slightly reduced levels of BAFF. In ET patients, we showed that the differentiation of CD34+ progenitor cells towards megakaryocytes induces the expression of both APRIL and BAFF. More importantly, APRIL neutralization significantly reduced platelet production. In conclusion, our data provide evidence that blocking APRIL signaling, which acts as an autocrine growth factor for terminal megakaryocytopoiesis, inhibits platelet production in ET patients, regardless of the status of JAK2(V617F) mutation.
Collapse
Affiliation(s)
- Lukasz Bolkun
- Department of Haematology, Medical University of Bialystok, ul. M. Skłodowskiej-Curie 24A, 15-276 Bialystok, Poland
- Correspondence: (L.B.); (A.E.); Tel.: +48-85-7468230 (L.B.); +48-85-748-59-72 (A.E.); Fax: +48-85-748-59-71 (A.E.)
| | - Marlena Tynecka
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269 Bialystok, Poland
| | - Tomasz Wasiluk
- Regional Centre for Transfusion Medicine, Bialystok, ul. M. Skłodowskiej-Curie 23, 15-950 Bialystok, Poland
| | - Jaroslaw Piszcz
- Department of Haematology, Medical University of Bialystok, ul. M. Skłodowskiej-Curie 24A, 15-276 Bialystok, Poland
| | - Aleksandra Starosz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269 Bialystok, Poland
| | - Kamil Grubczak
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269 Bialystok, Poland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269 Bialystok, Poland
- Department of Allergology and Internal Medicine, Medical University of Bialystok, ul. M. Skłodowskiej-Curie 24A, 15-276 Bialystok, Poland
| | - Andrzej Eljaszewicz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269 Bialystok, Poland
- Correspondence: (L.B.); (A.E.); Tel.: +48-85-7468230 (L.B.); +48-85-748-59-72 (A.E.); Fax: +48-85-748-59-71 (A.E.)
| |
Collapse
|
35
|
Frank D, Garnish SE, Sandow JJ, Weir A, Liu L, Clayer E, Meza L, Rashidi M, Cobbold SA, Scutts SR, Doerflinger M, Anderton H, Lawlor KE, Lalaoui N, Kueh AJ, Eng VV, Ambrose RL, Herold MJ, Samson AL, Feltham R, Murphy JM, Ebert G, Pearson JS, Vince JE. Ubiquitylation of RIPK3 beyond-the-RHIM can limit RIPK3 activity and cell death. iScience 2022; 25:104632. [PMID: 35800780 PMCID: PMC9254354 DOI: 10.1016/j.isci.2022.104632] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 03/31/2022] [Accepted: 06/13/2022] [Indexed: 12/05/2022] Open
Abstract
Pathogen recognition and TNF receptors signal via receptor interacting serine/threonine kinase-3 (RIPK3) to cause cell death, including MLKL-mediated necroptosis and caspase-8-dependent apoptosis. However, the post-translational control of RIPK3 is not fully understood. Using mass-spectrometry, we identified that RIPK3 is ubiquitylated on K469. The expression of mutant RIPK3 K469R demonstrated that RIPK3 ubiquitylation can limit both RIPK3-mediated apoptosis and necroptosis. The enhanced cell death of overexpressed RIPK3 K469R and activated endogenous RIPK3 correlated with an overall increase in RIPK3 ubiquitylation. Ripk3K469R/K469R mice challenged with Salmonella displayed enhanced bacterial loads and reduced serum IFNγ. However, Ripk3K469R/K469R macrophages and dermal fibroblasts were not sensitized to RIPK3-mediated apoptotic or necroptotic signaling suggesting that, in these cells, there is functional redundancy with alternate RIPK3 ubiquitin-modified sites. Consistent with this idea, the mutation of other ubiquitylated RIPK3 residues also increased RIPK3 hyper-ubiquitylation and cell death. Therefore, the targeted ubiquitylation of RIPK3 may act as either a brake or accelerator of RIPK3-dependent killing.
RIPK3 can be ubiquitylated on K469 to limit RIPK3-induced necroptosis and apoptosis Ripk3K469R/K469R mice are more susceptible to Salmonella infection Several ubiquitylated or surface exposed lysines can limit RIPK3-induced cell death Hyper-ubiquitylated RIPK3 correlates with RIPK3 signaling and cell death
Collapse
|
36
|
Meng Y, Horne CR, Samson AL, Dagley LF, Young SN, Sandow JJ, Czabotar PE, Murphy JM. Human RIPK3 C-lobe phosphorylation is essential for necroptotic signaling. Cell Death Dis 2022; 13:565. [PMID: 35739084 PMCID: PMC9226014 DOI: 10.1038/s41419-022-05009-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 01/21/2023]
Abstract
Necroptosis is a caspase-independent, pro-inflammatory mode of programmed cell death which relies on the activation of the terminal effector, MLKL, by the upstream protein kinase RIPK3. To mediate necroptosis, RIPK3 must stably interact with, and phosphorylate the pseudokinase domain of MLKL, although the precise molecular cues that provoke RIPK3 necroptotic signaling are incompletely understood. The recent finding that RIPK3 S227 phosphorylation and the occurrence of a stable RIPK3:MLKL complex in human cells prior to exposure to a necroptosis stimulus raises the possibility that additional, as-yet-unidentified phosphorylation events activate RIPK3 upon initiation of necroptosis signaling. Here, we sought to identify phosphorylation sites of RIPK3 and dissect their regulatory functions. Phosphoproteomics identified 21 phosphorylation sites in HT29 cells overexpressing human RIPK3. By comparing cells expressing wild-type and kinase-inactive D142N RIPK3, autophosphorylation sites and substrates of other cellular kinases were distinguished. Of these 21 phosphosites, mutational analyses identified only pT224 and pS227 as crucial, synergistic sites for stable interaction with MLKL to promote necroptosis, while the recently reported activation loop phosphorylation at S164/T165 negatively regulate the kinase activity of RIPK3. Despite being able to phosphorylate MLKL to a similar or higher extent than wild-type RIPK3, mutation of T224, S227, or the RHIM in RIPK3 attenuated necroptosis. This finding highlights the stable recruitment of human MLKL by RIPK3 to the necrosome as an essential checkpoint in necroptosis signaling, which is independent from and precedes the phosphorylation of MLKL.
Collapse
Affiliation(s)
- Yanxiang Meng
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Parkville, VIC 3052 Australia
| | - Christopher R. Horne
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Parkville, VIC 3052 Australia
| | - Andre L. Samson
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Parkville, VIC 3052 Australia
| | - Laura F. Dagley
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Parkville, VIC 3052 Australia
| | - Samuel N. Young
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052 Australia
| | - Jarrod J. Sandow
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Parkville, VIC 3052 Australia
| | - Peter E. Czabotar
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Parkville, VIC 3052 Australia
| | - James M. Murphy
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Parkville, VIC 3052 Australia
| |
Collapse
|
37
|
LTα, TNF, and ILC3 in Peyer's Patch Organogenesis. Cells 2022; 11:cells11121970. [PMID: 35741098 PMCID: PMC9221848 DOI: 10.3390/cells11121970] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/11/2022] [Accepted: 06/17/2022] [Indexed: 02/05/2023] Open
Abstract
TNF and LTα are structurally related cytokines of the TNF superfamily. Their genes are located in close proximity to each other and to the Ltb gene within the TNF/LT locus inside MHC. Unlike Ltb, transcription of Tnf and of Lta is tightly controlled, with the Tnf gene being an immediate early gene that is rapidly induced in response to various inflammatory stimuli. Genes of the TNF/LT locus play a crucial role in lymphoid tissue organogenesis, although some aspects of their specific contribution remain controversial. Here, we present new findings and discuss the distinct contribution of TNF produced by ILC3 cells to Peyer’s patch organogenesis.
Collapse
|
38
|
Lower Expression of TWEAK is Associated with Poor Survival and Dysregulate TIICs in Lung Adenocarcinoma. DISEASE MARKERS 2022; 2022:8661423. [PMID: 35707713 PMCID: PMC9192298 DOI: 10.1155/2022/8661423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 02/22/2022] [Accepted: 05/03/2022] [Indexed: 12/24/2022]
Abstract
Background. Lung cancer remains the leading cause of cancer death worldwide, and the most subtype is lung adenocarcinoma (LUAD). Tumor-infiltrating immune cells (TIICs) greatly impact the prognosis of LUAD. Tumor necrosis factor–like weak inducer of apoptosis (TWEAK), signal via its receptor fibroblast growth factor-inducible 14 (Fn14), dysregulates immune cell recruitment within tumor environment, thus promoting the progression of autoimmune diseases and cancer. We aimed to explore its role in LUAD. Methods. The expression level of TWEAK was explored in Tumor Immune Estimation Resource 2.0 (TIMER2.0) and Oncomine databases. The Tumor Immune Dysfunction and Exclusion (TIDE) and Lung Cancer Explorer (LCE) databases were applied to evaluate the survival in correlation to TWEAK expression. TIICs were assessed with TIMER2.0 and TIDE datasets. The expression of TWEAK protein was detected in LUAD cell lines and also in tissue samples from LUAD patients via western blotting or combination with immunochemistry. Results. Our results showed that TWEAK was downregulated in LUAD tumors compared to normal tissues in TIMER2.0, Oncomine, cell lines, and clinical specimens. Poor survival was uncovered in lower TWEAK expression of LUAD patients in LCE (
[95% CI, 0.76-0.92]) and TCGA (
,
) and GSE13213@PRECOG (
,
) in TIDE. Multiple tumor-infiltrating immune cells (TIICs) were found closely correlated with TWEAK expression in LUAD, especially hematopoietic stem cell (
,
), common lymphoid progenitor (
,
), and myeloid-derived suppressor cells (MDSCs) (
,
). Conclusion. Lower level of TWEAK was linked with poor survival and aberrant recruitment and phenotype of TIICs in LUAD, which might motivate immune escape and weaken the effects of immunotherapy.
Collapse
|
39
|
The Lck inhibitor, AMG-47a, blocks necroptosis and implicates RIPK1 in signalling downstream of MLKL. Cell Death Dis 2022; 13:291. [PMID: 35365636 PMCID: PMC8976052 DOI: 10.1038/s41419-022-04740-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 12/11/2022]
Abstract
Necroptosis is a form of caspase-independent programmed cell death that arises from disruption of cell membranes by the mixed lineage kinase domain-like (MLKL) pseudokinase after its activation by the upstream kinases, receptor interacting protein kinase (RIPK)-1 and RIPK3, within a complex known as the necrosome. Dysregulated necroptosis has been implicated in numerous inflammatory pathologies. As such, new small molecule necroptosis inhibitors are of great interest, particularly ones that operate downstream of MLKL activation, where the pathway is less well defined. To better understand the mechanisms involved in necroptosis downstream of MLKL activation, and potentially uncover new targets for inhibition, we screened known kinase inhibitors against an activated mouse MLKL mutant, leading us to identify the lymphocyte-specific protein tyrosine kinase (Lck) inhibitor AMG-47a as an inhibitor of necroptosis. We show that AMG-47a interacts with both RIPK1 and RIPK3, that its ability to protect from cell death is dependent on the strength of the necroptotic stimulus, and that it blocks necroptosis most effectively in human cells. Moreover, in human cell lines, we demonstrate that AMG-47a can protect against cell death caused by forced dimerisation of MLKL truncation mutants in the absence of any upstream signalling, validating that it targets a process downstream of MLKL activation. Surprisingly, however, we also found that the cell death driven by activated MLKL in this model was completely dependent on the presence of RIPK1, and to a lesser extent RIPK3, although it was not affected by known inhibitors of these kinases. Together, these results suggest an additional role for RIPK1, or the necrosome, in mediating human necroptosis after MLKL is phosphorylated by RIPK3 and provide further insight into reported differences in the progression of necroptosis between mouse and human cells.
Collapse
|
40
|
Ninnemann J, Winsauer C, Bondareva M, Kühl AA, Lozza L, Durek P, Lissner D, Siegmund B, Kaufmann SHE, Mashreghi MF, Nedospasov SA, Kruglov AA. TNF hampers intestinal tissue repair in colitis by restricting IL-22 bioavailability. Mucosal Immunol 2022; 15:698-716. [PMID: 35383266 PMCID: PMC9259490 DOI: 10.1038/s41385-022-00506-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/10/2022] [Indexed: 02/04/2023]
Abstract
Successful treatment of chronic inflammatory diseases integrates both the cessation of inflammation and the induction of adequate tissue repair processes. Strikingly, targeting a single proinflammatory cytokine, tumor necrosis factor (TNF), induces both processes in a relevant cohort of inflammatory bowel disease (IBD) patients. However, the molecular mechanisms underlying intestinal repair following TNF blockade during IBD remain elusive. Using a novel humanized model of experimental colitis, we demonstrate that TNF interfered with the tissue repair program via induction of a soluble natural antagonist of IL-22 (IL-22Ra2; IL-22BP) in the colon and abrogated IL-22/STAT3-mediated mucosal repair during colitis. Furthermore, membrane-bound TNF expressed by T cells perpetuated colonic inflammation, while soluble TNF produced by epithelial cells (IECs) induced IL-22BP expression in colonic dendritic cells (DCs) and dampened IL-22-driven restitution of colonic epithelial functions. Finally, TNF induced IL-22BP expression in human monocyte-derived DCs and levels of IL22-BP correlated with TNF in sera of IBD patients. Thus, our data can explain how anti-TNF therapy induces mucosal healing by increasing IL-22 availability and implicates new therapeutic opportunities for IBD.
Collapse
Affiliation(s)
- Justus Ninnemann
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Caroline Winsauer
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Marina Bondareva
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
- Belozersky Institute of Physico-Chemical Biology and Faculty of Bioengineering and Bioinformatics, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Anja A Kühl
- iPATH.Berlin, Core Unit of Charité-Universitätsmedizin Berlin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Laura Lozza
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Pawel Durek
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Donata Lissner
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan H E Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | | | - Sergei A Nedospasov
- Belozersky Institute of Physico-Chemical Biology and Faculty of Bioengineering and Bioinformatics, M.V. Lomonosov Moscow State University, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andrey A Kruglov
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany.
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
41
|
Rusu I, Mennillo E, Bain JL, Li Z, Sun X, Ly KM, Rosli YY, Naser M, Wang Z, Advincula R, Achacoso P, Shao L, Razani B, Klein OD, Marson A, Turnbaugh JA, Turnbaugh PJ, Malynn BA, Ma A, Kattah MG. Microbial signals, MyD88, and lymphotoxin drive TNF-independent intestinal epithelial tissue damage. J Clin Invest 2022; 132:154993. [PMID: 35077396 PMCID: PMC8884902 DOI: 10.1172/jci154993] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/19/2022] [Indexed: 11/18/2022] Open
Abstract
Anti-TNF antibodies are effective for treating patients with inflammatory bowel disease (IBD), but many patients fail to respond to anti-TNF therapy, highlighting the importance of TNF-independent disease. We previously demonstrated that acute deletion of 2 IBD susceptibility genes, A20 (Tnfaip3) and Abin-1 (Tnip1), in intestinal epithelial cells (IECs) sensitized mice to both TNF-dependent and TNF-independent death. Here we show that TNF-independent IEC death after A20 and Abin-1 deletion was rescued by germ-free derivation or deletion of MyD88, while deletion of Trif provided only partial protection. Combined deletion of Ripk3 and Casp8, which inhibits both apoptotic and necroptotic death, completely protected against death after acute deletion of A20 and Abin-1 in IECs. A20- and Abin-1–deficient IECs were sensitized to TNF-independent, TNFR1-mediated death in response to lymphotoxin α (LTα) homotrimers. Blockade of LTα in vivo reduced weight loss and improved survival when combined with partial deletion of MyD88. Biopsies of inflamed colon mucosa from patients with IBD exhibited increased LTA and IL1B expression, including a subset of patients with active colitis on anti-TNF therapy. These data show that microbial signals, MyD88, and LTα all contribute to TNF-independent intestinal injury.
Collapse
Affiliation(s)
- Iulia Rusu
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Elvira Mennillo
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Jared L. Bain
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Zhongmei Li
- Department of Medicine, UCSF, San Francisco, California, USA
- Gladstone Institutes, San Francisco, California, USA
| | - Xiaofei Sun
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Yenny Y. Rosli
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Mohammad Naser
- Biological Imaging Development CoLab, UCSF, San Francisco, California, USA
| | - Zunqiu Wang
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Philip Achacoso
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Ling Shao
- Department of Medicine, University of Southern California, Los Angeles, California, USA
| | | | - Ophir D. Klein
- Departments of Orofacial Sciences and Pediatrics, Program in Craniofacial Biology, and
| | - Alexander Marson
- Department of Medicine, UCSF, San Francisco, California, USA
- Gladstone Institutes, San Francisco, California, USA
- Department of Microbiology and Immunology and
- Institute for Human Genetics, UCSF, San Francisco, California, USA
- Innovative Genomics Institute, University of California, Berkeley, California, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | | | | | | | - Averil Ma
- Department of Medicine, UCSF, San Francisco, California, USA
| | | |
Collapse
|
42
|
Karolin A, Escher G, Rudloff S, Sidler D. Nephrotoxicity of Calcineurin Inhibitors in Kidney Epithelial Cells is Independent of NFAT Signaling. Front Pharmacol 2022; 12:789080. [PMID: 35140605 PMCID: PMC8819135 DOI: 10.3389/fphar.2021.789080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/29/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Calcineurin inhibitors (CNIs) such as cyclosporine A and tacrolimus are commonly used after renal transplantation to suppress the immune system. In lymphoid cells, cyclosporine A acts via the calcineurin/nuclear factor of activated T-cell (NFAT) axis. In non-lymphoid cells, such as kidney epithelial cells, cyclosporine A induces calcineurin inhibitor toxicity. It is unknown via which off-targets cyclosporine A induces calcineurin inhibitor toxicity in kidney epithelial cells. Methods: To measure a compound’s potential to induce nephrotoxicity, the expression of the surrogate marker Fn14 was measured by flow cytometry. Compounds were tested for their potential to induce Fn14 either chemically or plasmid-mediated. Mice were injected with various compounds, and changes in nephrotoxic gene expression levels of the kidney epithelial cells were then analyzed. Results: Fn14 is specifically upregulated due to calcineurin inhibitor toxicity inducing agents. Inhibition of the NFAT axis showed no increase of the Fn14 expression on the surface of kidney cells. However, inhibition of p38 MAPK, phosphoinositide-3-kinase (PI3K)/Akt, protein kinase C (PKC), and inhibitor of nuclear factor-κB (IκB) kinase (IKK) showed clear induction of Fn14 and increased expressions of nephrotoxic, inflammatory, and fibrotic genes in vitro and in vivo. Conclusions: These findings show that cyclosporine A acts independently of NFAT on kidney epithelial cells. Moreover, inhibition of serine/threonine protein kinases mimics cyclosporine A’s activity on kidney epithelial cells. This mimicking effect indicates that these protein kinases are off-targets of cyclosporine A and damage structural renal cells when inhibited and therefore contributes likely to the development and progression of calcineurin inhibitor toxicity.
Collapse
Affiliation(s)
- Andrea Karolin
- Department for Nephrology and Hypertension, University Hospital Insel Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Geneviève Escher
- Department for Nephrology and Hypertension, University Hospital Insel Bern, Bern, Switzerland
| | - Stefan Rudloff
- Department for Nephrology and Hypertension, University Hospital Insel Bern, Bern, Switzerland
| | - Daniel Sidler
- Department for Nephrology and Hypertension, University Hospital Insel Bern, Bern, Switzerland
- *Correspondence: Daniel Sidler,
| |
Collapse
|
43
|
The deubiquitinase Usp27x as a novel regulator of cFLIP L protein expression and sensitizer to death-receptor-induced apoptosis. Apoptosis 2022; 27:112-132. [PMID: 35044632 PMCID: PMC8863773 DOI: 10.1007/s10495-021-01706-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 11/02/2022]
Abstract
Death receptors are transmembrane proteins that can induce the activation of caspase-8 upon ligand binding, initiating apoptosis. Recent work has highlighted the great molecular complexity of death receptor signalling, in particular through ubiquitination/deubiquitination. We have earlier defined the deubiquitinase Ubiquitin-Specific Protease 27x (Usp27x) as an enzyme capable of stabilizing the pro-apoptotic Bcl-2 family member Bim. Here, we report that enhanced expression of Usp27x in human melanoma cells leads to the loss of cellular FLICE-like inhibitory protein (cFLIP) and sensitizes to Tumor necrosis factor receptor 1 (TNF-R1) or Toll-like receptor 3 (TLR3)-induced extrinsic apoptosis through enabling enhanced processing of caspase-8. The loss of cFLIPL upon overexpression of Usp27x was not due to reduced transcription, could be partially counteracted by blocking the ubiquitin proteasome system and was independent of the known cFLIPL destabilizing ubiquitin E3-ligases Itch and DTX1. Instead, Usp27x interacted with the E3-ligase TRIM28 and reduced ubiquitination of TRIM28. Reduction of cFLIPL protein levels by Usp27x-induction depended on TRIM28, which was also required for polyI:C-induced cell death. This work defines Usp27x as a novel regulator of cFLIPL protein expression and a deubiquitinase in fine tuning death receptor signalling pathways to execute apoptosis.
Collapse
|
44
|
Liu W, Chou TF, Garrett-Thomson SC, Seo GY, Fedorov E, Ramagopal UA, Bonanno JB, Wang Q, Kim K, Garforth SJ, Kakugawa K, Cheroutre H, Kronenberg M, Almo SC. HVEM structures and mutants reveal distinct functions of binding to LIGHT and BTLA/CD160. J Exp Med 2021; 218:e20211112. [PMID: 34709351 PMCID: PMC8558838 DOI: 10.1084/jem.20211112] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/20/2021] [Accepted: 10/01/2021] [Indexed: 11/09/2022] Open
Abstract
HVEM is a TNF (tumor necrosis factor) receptor contributing to a broad range of immune functions involving diverse cell types. It interacts with a TNF ligand, LIGHT, and immunoglobulin (Ig) superfamily members BTLA and CD160. Assessing the functional impact of HVEM binding to specific ligands in different settings has been complicated by the multiple interactions of HVEM and HVEM binding partners. To dissect the molecular basis for multiple functions, we determined crystal structures that reveal the distinct HVEM surfaces that engage LIGHT or BTLA/CD160, including the human HVEM-LIGHT-CD160 ternary complex, with HVEM interacting simultaneously with both binding partners. Based on these structures, we generated mouse HVEM mutants that selectively recognized either the TNF or Ig ligands in vitro. Knockin mice expressing these muteins maintain expression of all the proteins in the HVEM network, yet they demonstrate selective functions for LIGHT in the clearance of bacteria in the intestine and for the Ig ligands in the amelioration of liver inflammation.
Collapse
MESH Headings
- Animals
- Antigens, CD/chemistry
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Crystallography, X-Ray
- Drosophila/cytology
- Drosophila/genetics
- Female
- GPI-Linked Proteins/chemistry
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Transgenic
- Multiprotein Complexes/chemistry
- Multiprotein Complexes/metabolism
- Mutation
- Receptors, Immunologic/chemistry
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, Tumor Necrosis Factor, Member 14/chemistry
- Receptors, Tumor Necrosis Factor, Member 14/genetics
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Tumor Necrosis Factor Ligand Superfamily Member 14/chemistry
- Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
- Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
- Yersinia Infections/genetics
- Yersinia Infections/pathology
- Mice
Collapse
Affiliation(s)
- Weifeng Liu
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY
| | | | | | | | - Elena Fedorov
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY
| | - Udupi A. Ramagopal
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY
| | - Jeffrey B. Bonanno
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY
| | | | - Kenneth Kim
- La Jolla Institute for Immunology, La Jolla, CA
| | - Scott J. Garforth
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY
| | - Kiyokazu Kakugawa
- Laboratory for Immune Crosstalk, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hilde Cheroutre
- La Jolla Institute for Immunology, La Jolla, CA
- Laboratory for Immune Crosstalk, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA
- Division of Biological Sciences, University of California San Diego, La Jolla, CA
| | - Steven C. Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
45
|
Van den Broek B, Wuyts C, Irobi J. Extracellular vesicle-associated small heat shock proteins as therapeutic agents in neurodegenerative diseases and beyond. Adv Drug Deliv Rev 2021; 179:114009. [PMID: 34673130 DOI: 10.1016/j.addr.2021.114009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/11/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022]
Abstract
Increasing evidence points towards using extracellular vesicles (EVs) as a therapeutic strategy in neurodegenerative diseases such as multiple sclerosis, Parkinson's, and Alzheimer's disease. EVs are nanosized carriers that play an essential role in intercellular communication and cellular homeostasis by transporting an active molecular cargo, including a large variety of proteins. Recent publications demonstrate that small heat shock proteins (HSPBs) exhibit a beneficial role in neurodegenerative diseases. Moreover, it is defined that HSPBs target the autophagy and the apoptosis pathway, playing a prominent role in chaperone activity and cell survival. This review elaborates on the therapeutic potential of EVs and HSPBs, in particular HSPB1 and HSPB8, in neurodegenerative diseases. We conclude that EVs and HSPBs positively influence neuroinflammation, central nervous system (CNS) repair, and protein aggregation in CNS disorders. Moreover, we propose the use of HSPB-loaded EVs as advanced nanocarriers for the future development of neurodegenerative disease therapies.
Collapse
Affiliation(s)
- Bram Van den Broek
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Charlotte Wuyts
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Joy Irobi
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium.
| |
Collapse
|
46
|
Meng Y, Davies KA, Fitzgibbon C, Young SN, Garnish SE, Horne CR, Luo C, Garnier JM, Liang LY, Cowan AD, Samson AL, Lessene G, Sandow JJ, Czabotar PE, Murphy JM. Human RIPK3 maintains MLKL in an inactive conformation prior to cell death by necroptosis. Nat Commun 2021; 12:6783. [PMID: 34811356 PMCID: PMC8608796 DOI: 10.1038/s41467-021-27032-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/29/2021] [Indexed: 12/11/2022] Open
Abstract
The ancestral origins of the lytic cell death mode, necroptosis, lie in host defense. However, the dysregulation of necroptosis in inflammatory diseases has led to widespread interest in targeting the pathway therapeutically. This mode of cell death is executed by the terminal effector, the MLKL pseudokinase, which is licensed to kill following phosphorylation by its upstream regulator, RIPK3 kinase. The precise molecular details underlying MLKL activation are still emerging and, intriguingly, appear to mechanistically-diverge between species. Here, we report the structure of the human RIPK3 kinase domain alone and in complex with the MLKL pseudokinase. These structures reveal how human RIPK3 structurally differs from its mouse counterpart, and how human RIPK3 maintains MLKL in an inactive conformation prior to induction of necroptosis. Residues within the RIPK3:MLKL C-lobe interface are crucial to complex assembly and necroptotic signaling in human cells, thereby rationalizing the strict species specificity governing RIPK3 activation of MLKL.
Collapse
Affiliation(s)
- Yanxiang Meng
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Katherine A Davies
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Cheree Fitzgibbon
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Samuel N Young
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Sarah E Garnish
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Christopher R Horne
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Cindy Luo
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Jean-Marc Garnier
- SYNthesis med chem, 30 Flemington Rd, Parkville, VIC, 3052, Australia
| | - Lung-Yu Liang
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Angus D Cowan
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Andre L Samson
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Guillaume Lessene
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Jarrod J Sandow
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Peter E Czabotar
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
| |
Collapse
|
47
|
Feoktistova M, Makarov R, Yazdi AS, Panayotova-Dimitrova D. RIPK1 and TRADD Regulate TNF-Induced Signaling and Ripoptosome Formation. Int J Mol Sci 2021; 22:ijms222212459. [PMID: 34830347 PMCID: PMC8617695 DOI: 10.3390/ijms222212459] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 11/16/2022] Open
Abstract
TNF is a proinflammatory cytokine that is critical for the coordination of tissue homeostasis. RIPK1 and TRADD are the main participants in the transduction of TNF signaling. However, data on the cell fate-controlling functions of both molecules are quite controversial. Here, we address the functions of RIPK1 and TRADD in TNF signaling by generating RIPK1- or TRADD-deficient human cell lines. We demonstrate that RIPK1 is relevant for TNF-induced apoptosis and necroptosis in conditions with depleted IAPs. In addition, TRADD is dispensable for necroptosis but required for apoptosis. We reveal a new possible function of TRADD as a negative regulator of NIK stabilization and subsequent ripoptosome formation. Furthermore, we show that RIPK1 and TRADD do not appear to be essential for the activation of MAPK signaling. Moreover, partially repressing NF-κB activation in both RIPK1 and TRADD KO cells does not result in sensitization to TNF alone due to the absence of NIK stabilization. Importantly, we demonstrate that RIPK1 is essential for preventing TRADD from undergoing TNF-induced ubiquitination and degradation. Taken together, our findings provide further insights into the specific functions of RIPK1 and TRADD in the regulation of TNF-dependent signaling, which controls the balance between cell death and survival.
Collapse
|
48
|
Tezuka Y, Eguchi-Ishimae M, Ozaki E, Ito T, Ishii E, Eguchi M. Activation of fibroblast growth factor-inducible 14 in the early phase of childhood IgA nephropathy. PLoS One 2021; 16:e0258090. [PMID: 34597335 PMCID: PMC8486145 DOI: 10.1371/journal.pone.0258090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 09/17/2021] [Indexed: 11/23/2022] Open
Abstract
IgA nephropathy (IgAN) is the most common form of glomerulonephritis worldwide. Pediatric patients in Japan are diagnosed with IgAN at an early stage of the disease through annual urinary examinations. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and fibroblast growth factor-inducible 14 (Fn14) have various roles, including proinflammatory effects, and modulation of several kidney diseases; however, no reports have described their roles in pediatric IgAN. In this study, we performed pathological and immunohistochemical analyses of samples from 14 pediatric IgAN patients. Additionally, gene expression arrays of glomeruli by laser-captured microdissection were performed in hemi-nephrectomized high serum IgA (HIGA) mice, a model of IgA nephropathy, to determine the role of Fn14. Glomeruli with intense Fn14 deposition were observed in 80% of mild IgAN cases; however, most severe cases showed glomeruli with little or no Fn14 deposition. Fn14 deposition was not observed in obvious mesangial proliferation or the crescent region of glomeruli, but was detected strongly in the glomerular tuft, with an intact appearance. In HIGA mice, Fn14 deposition was observed mildly beginning at 11 weeks of age, and stronger Fn14 deposition was detected at 14 weeks of age. Expression array analysis indicated that Fn14 expression was higher in HIGA mice at 6 weeks of age, increased slightly at 11 weeks, and then decreased at 26 weeks when compared with controls at equivalent ages. These findings suggest that Fn14 signaling affects early lesions but not advanced lesions in patients with IgAN. Further study of the TWEAK/Fn14 pathway will contribute to our understanding of the progression of IgAN.
Collapse
Affiliation(s)
- Yuko Tezuka
- Department of Pediatrics, Takamatsu Red Cross Hospital, Takamatsu, Kagawa, Japan
| | | | - Erina Ozaki
- Department of Total Medical Support Center, Ehime University Hospital, Toon, Ehime, Japan
| | - Toshiyuki Ito
- Department of Pediatrics, Takamatsu Red Cross Hospital, Takamatsu, Kagawa, Japan
| | - Eiichi Ishii
- Department of Pediatrics, Takamatsu Red Cross Hospital, Takamatsu, Kagawa, Japan
| | - Mariko Eguchi
- Department of Pediatrics, Takamatsu Red Cross Hospital, Takamatsu, Kagawa, Japan
- Division of Medical Genetics, Ehime University Hospital, Toon, Ehime, Japan
- * E-mail:
| |
Collapse
|
49
|
Zhao M, Fu L, Chai Y, Sun M, Li Y, Wang S, Qi J, Zeng B, Kang L, Gao GF, Tan S. Atypical TNF-TNFR superfamily binding interface in the GITR-GITRL complex for T cell activation. Cell Rep 2021; 36:109734. [PMID: 34551288 DOI: 10.1016/j.celrep.2021.109734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/16/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022] Open
Abstract
Glucocorticoid-induced tumor necrosis factor receptor family-related protein (GITR) is a critical regulatory molecule in modulation of T cell immune responses. Here we report the mouse GITR (mGITR) and mGITR ligand (mGITRL) complex structure and find that the binding interface of mGITR and mGITRL is distinct from the typical tumor necrosis factor superfamily (TNFSF)/TNF receptor superfamily (TNFRSF) members. mGITR binds to its ligand with a single domain, whereas the binding interface on mGITRL is located on the side, which is distal from conserved binding sites of TNFSF molecules. Mutational analysis reveals that the binding interface of GITR/GITRL in humans is conserved with that in the mouse. Substitution of key interacting D93-I94-V95 (DIV) in mGITR with the corresponding K93-F94-S95 (KFS) in human GITR enables cross-recognition with human GITRL and cross-activation of receptor signaling. The findings of this study substantially expand our understanding of the interaction of TNFSF/TNFRSF superfamily molecules and can benefit the future design of biologics by targeting GITR/GITRL.
Collapse
Affiliation(s)
- Min Zhao
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Lijun Fu
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Yan Chai
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Meng Sun
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Li
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Shuo Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianxun Qi
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bin Zeng
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China; College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Le Kang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - George F Gao
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Shuguang Tan
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
50
|
Dakhel S, Lizak C, Matasci M, Mock J, Villa A, Neri D, Cazzamalli S. An Attenuated Targeted-TNF Localizes to Tumors In Vivo and Regains Activity at the Site of Disease. Int J Mol Sci 2021; 22:10020. [PMID: 34576184 PMCID: PMC8469155 DOI: 10.3390/ijms221810020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 11/16/2022] Open
Abstract
Antibody-cytokine fusion proteins (immunocytokines) are gaining importance for cancer therapy, but those products are often limited by systemic toxicity related to the activity of the cytokine payload in circulation and in secondary lymphoid organs. Tumor necrosis factor (TNF) is used as a pro-inflammatory payload to trigger haemorrhagic necrosis and boost anti-cancer immunity at the tumor site. Here we describe a depotentiated version of TNF (carrying the single point mutation I97A), which displayed reduced binding affinity to its cognate receptor tumor necrosis factor receptor 1 (TNFR-1) and lower biocidal activity. The fusion of the TNF(I97A) mutant to the L19 antibody promoted restoration of anti-tumor activity upon accumulation on the cognate antigen, the alternatively spliced EDB domain of fibronectin. In vivo administration of high doses (375 μg/Kg) of the fusion protein showed a potent anti-tumor effect without apparent toxicity compared with the wild type protein. L19-TNFI97A holds promise for the targeted delivery of TNF activity to neoplastic lesions, helping spare normal tissues.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal, Humanized/genetics
- Antibodies, Monoclonal, Humanized/metabolism
- Cricetulus
- Cytokines/genetics
- Cytokines/metabolism
- Female
- Fibronectins/genetics
- Fibronectins/metabolism
- Fluorescent Antibody Technique
- Immunotherapy
- Mice, Inbred BALB C
- Mutation
- Protein Structure, Secondary
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
- Mice
Collapse
Affiliation(s)
- Sheila Dakhel
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
| | - Christian Lizak
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
| | - Mattia Matasci
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
| | - Jacqueline Mock
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
| | - Alessandra Villa
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
| | - Dario Neri
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
- Philogen S.p.A., Piazza La Lizza, 7, 53100 Siena, Italy
| | - Samuele Cazzamalli
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
| |
Collapse
|