1
|
Chattopadhyay S, Hazra R, Mallick A, Gayen S, Roy S. Small-molecule in cancer immunotherapy: Revolutionizing cancer treatment with transformative, game-changing breakthroughs. Biochim Biophys Acta Rev Cancer 2024; 1879:189170. [PMID: 39127244 DOI: 10.1016/j.bbcan.2024.189170] [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: 06/13/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Immunotherapy has revolutionized cancer management, with antibody-based treatments leading the charge due to their superior pharmacodynamics, including enhanced effectiveness and specificity. However, these therapies are hampered by limitations such as prolonged half-lives, poor tissue and tumor penetration, and minimal oral bioavailability. Additionally, their immunogenic nature can cause adverse effects. Consequently, the focus is shifting towards small-molecule-based immunotherapies, which potentially overcome these drawbacks. Emerging as a promising alternative, small molecules offer the benefits of therapeutic antibodies and immunomodulators, often yielding synergistic effects when combined. Recent advancements in small-molecule cancer immunotherapy are notable, featuring inhibitors, agonists, and degraders that act as immunomodulators. This article delves into the current landscape of small-molecule immunotherapy in cancer treatment, highlighting novel agents targeting key pathways such as Toll-like receptors (TLR), PD-1/PD-L1, chemokine receptors, and stimulators of interferon genes (STING). The review emphasizes newly discovered molecular entities and their modulatory roles in tumorigenesis, many of which have progressed to clinical trials, that aims to provide a comprehensive snapshot of the evolving frontier in cancer treatment, driven by small-molecule immunomodulators.
Collapse
Affiliation(s)
- Soumyadeep Chattopadhyay
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India
| | - Rudradeep Hazra
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India
| | - Arijit Mallick
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India
| | - Sakuntala Gayen
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India.
| |
Collapse
|
2
|
Lu G, Liu H, Wang H, Tang X, Luo S, Du M, Christiani DC, Wei Q. Potentially functional variants of INPP5D and EXOSC3 in immunity B cell-related genes are associated with non-small cell lung cancer survival. Front Immunol 2024; 15:1440454. [PMID: 39176091 PMCID: PMC11338758 DOI: 10.3389/fimmu.2024.1440454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
B cells are adaptive immune cells in the tumor microenvironment and play an important role in tumor development and metastasis. However, the roles of genetic variants of the immunity B cell-related genes in the survival of patients with non-small cell lung cancer (NSCLC) remain unknown. In the present study, we first evaluated associations between 10,776 single nucleotide polymorphisms (SNPs) in 220 immunity B cell-related genes and survival of NSCLC in a discovery dataset of 1,185 patients from the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial. We found that 369 SNPs were significantly associated with overall survival (OS) of NSCLC in multivariable Cox proportional hazards regression analysis (P ≤ 0.05, Bayesian false discovery probability ≤ 0.80), of which 18 SNPs were validated in another independent genotyping dataset of 984 patients from the Harvard Lung Cancer Susceptibility (HLCS) Study. We then performed linkage disequilibrium (LD) analysis, followed by stepwise analysis with a multivariable Cox regression model. Finally, two independent SNPs, inositol polyphosphate-5-phosphatase D (INPP5D) rs13385922 C>T and exosome component 3 (EXOSC3) rs3208406 A>G, remained significantly associated withNSCLC OS with a combined hazards ratio (HR) of 1.14 (95% confidence interval = 1.06-1.23, P = 2.41×10-4) and 1.20 (95% confidence interval = 1.14-1.28, P = 3.41×10-9), respectively. Furthermore, NSCLC patients with the combination of unfavorable genotypes for these two SNPs were associated with a poor OS (P trend = 0.0002) and disease-specific survival (DSS, P trend < 0.0001) in the PLCO dataset. Expression quantitative trait loci (eQTL) analysis suggested that the INPP5D rs6782875 T allele was significantly correlated with elevated INPP5D mRNA expression levels in normal lung tissues and whole blood samples, while the EXOSC3 rs3208406 G allele was significantly correlated with increased EXOSC3 mRNA expression levels in normal lung tissues. Our data indicated that genetic variants in these immunity B cell-related genes may predict NSCLC survival possibly by influencing the gene expression.
Collapse
Affiliation(s)
- Guojun Lu
- Department of Respiratory Medicine, Nanjing Chest Hospital, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, United States
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, United States
| | - Huilin Wang
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, United States
- Department of Respiratory Oncology, Guangxi Cancer Hospital, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaozhun Tang
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, United States
- Department of Head and Neck Surgery, Guangxi Cancer Hospital, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, United States
| | - Mulong Du
- Departments of Environmental Health and Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, United States
| | - David C. Christiani
- Departments of Environmental Health and Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, United States
- Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, United States
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Duke Global Health Institute, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
3
|
Chou V, Pearse RV, Aylward AJ, Ashour N, Taga M, Terzioglu G, Fujita M, Fancher SB, Sigalov A, Benoit CR, Lee H, Lam M, Seyfried NT, Bennett DA, De Jager PL, Menon V, Young-Pearse TL. INPP5D regulates inflammasome activation in human microglia. Nat Commun 2023; 14:7552. [PMID: 38016942 PMCID: PMC10684891 DOI: 10.1038/s41467-023-42819-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/20/2023] [Indexed: 11/30/2023] Open
Abstract
Microglia and neuroinflammation play an important role in the development and progression of Alzheimer's disease (AD). Inositol polyphosphate-5-phosphatase D (INPP5D/SHIP1) is a myeloid-expressed gene genetically-associated with AD. Through unbiased analyses of RNA and protein profiles in INPP5D-disrupted iPSC-derived human microglia, we find that reduction in INPP5D activity is associated with molecular profiles consistent with disrupted autophagy and inflammasome activation. These findings are validated through targeted pharmacological experiments which demonstrate that reduced INPP5D activity induces the formation of the NLRP3 inflammasome, cleavage of CASP1, and secretion of IL-1β and IL-18. Further, in-depth analyses of human brain tissue across hundreds of individuals using a multi-analytic approach provides evidence that a reduction in function of INPP5D in microglia results in inflammasome activation in AD. These findings provide insights into the molecular mechanisms underlying microglia-mediated processes in AD and highlight the inflammasome as a potential therapeutic target for modulating INPP5D-mediated vulnerability to AD.
Collapse
Affiliation(s)
- Vicky Chou
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Richard V Pearse
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Aimee J Aylward
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Nancy Ashour
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Mariko Taga
- Center for Translational and Computational Neuroimmunology, Department of Neurology, and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Gizem Terzioglu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Masashi Fujita
- Center for Translational and Computational Neuroimmunology, Department of Neurology, and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Seeley B Fancher
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alina Sigalov
- Center for Translational and Computational Neuroimmunology, Department of Neurology, and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Courtney R Benoit
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hyo Lee
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Matti Lam
- Center for Translational and Computational Neuroimmunology, Department of Neurology, and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Vilas Menon
- Center for Translational and Computational Neuroimmunology, Department of Neurology, and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
4
|
Chowdhury BP, Das S, Bodhale N, Prakash Pandey S, Sudan R, Srivastava N, Chisholm JD, Kerr WG, Majumdar S, Saha B. SHIP1 inhibition via 3-alpha-amino-cholestane enhances protection against Leishmania infection. Cytokine 2023; 171:156373. [PMID: 37776719 DOI: 10.1016/j.cyto.2023.156373] [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: 07/26/2023] [Revised: 09/12/2023] [Accepted: 09/16/2023] [Indexed: 10/02/2023]
Abstract
Leishmania major and L. donovani cause cutaneous leishmaniasis and visceral leishmaniasis, respectively. Available chemotherapies suffer from toxicity, drug-resistance or high cost of production prompting the need for the discovery of new anti-leishmanials. Here, we test a novel aminosteriodal compound- 3-alpha-amino-cholestane [3AC] - that shows selective inhibition of SHIP1, an inositol-5'-phosphate-specific phosphatase with potent effects on the immune system. We report that 3AC-sensitive SHIP1 expression increases in Leishmania-infected macrophages. Treatment of BALB/c mice, a Leishmania-susceptible host, with 3AC increased anti-leishmanial, but reduced pro-leishmanial, cytokines' production and reduced the parasite load in both L. major and L. donovani infections. These findings implicate SHIPi as a potential novel immunostimulant with anti-leishmanial function.
Collapse
Affiliation(s)
| | - Shibali Das
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | | | - Raki Sudan
- Dept. of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Neetu Srivastava
- Dept. of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - John D Chisholm
- Dept. of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - William G Kerr
- Dept. of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA; Dept. of Chemistry, Syracuse University, Syracuse, NY, USA; Dept. of Pediatrics, SUNY Upstate Medical University, Syracuse, NY, USA.
| | | | | |
Collapse
|
5
|
Chou V, Fancher SB, Pearse RV, Lee H, Lam M, Seyfried NT, Bennett DA, De Jager PL, Menon V, Young-Pearse TL. INPP5D/SHIP1 regulates inflammasome activation in human microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.25.530025. [PMID: 36865139 PMCID: PMC9980181 DOI: 10.1101/2023.02.25.530025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Microglia and neuroinflammation are implicated in the development and progression of Alzheimer's disease (AD). To better understand microglia-mediated processes in AD, we studied the function of INPP5D/SHIP1, a gene linked to AD through GWAS. Immunostaining and single nucleus RNA sequencing confirmed that INPP5D expression in the adult human brain is largely restricted to microglia. Examination of prefrontal cortex across a large cohort revealed reduced full length INPP5D protein levels in AD patient brains compared to cognitively normal controls. The functional consequences of reduced INPP5D activity were evaluated in human induced pluripotent stem cell derived microglia (iMGLs), using both pharmacological inhibition of the phosphatase activity of INPP5D and genetic reduction in copy number. Unbiased transcriptional and proteomic profiling of iMGLs suggested an upregulation of innate immune signaling pathways, lower levels of scavenger receptors, and altered inflammasome signaling with INPP5D reduction. INPP5D inhibition induced the secretion of IL-1ß and IL-18, further implicating inflammasome activation. Inflammasome activation was confirmed through visualization of inflammasome formation through ASC immunostaining in INPP5D-inhibited iMGLs, increased cleaved caspase-1 and through rescue of elevated IL-1ß and IL-18 with caspase-1 and NLRP3 inhibitors. This work implicates INPP5D as a regulator of inflammasome signaling in human microglia.
Collapse
|
6
|
Fernandes S, Srivastava N, Pedicone C, Sudan R, Luke EA, Dungan OM, Pacherille A, Meyer ST, Dormann S, Schurmans S, Chambers BJ, Chisholm JD, Kerr WG. Obesity control by SHIP inhibition requires pan-paralog inhibition and an intact eosinophil compartment. iScience 2023; 26:106071. [PMID: 36818285 PMCID: PMC9929608 DOI: 10.1016/j.isci.2023.106071] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/18/2022] [Accepted: 01/23/2023] [Indexed: 01/29/2023] Open
Abstract
Here we extend the understanding of how chemical inhibition of SHIP paralogs controls obesity. We compare different classes of SHIP inhibitors and find that selective inhibitors of SHIP1 or SHIP2 are unable to prevent weight gain and body fat accumulation during increased caloric intake. Surprisingly, only pan-SHIP1/2 inhibitors (pan-SHIPi) prevent diet-induced obesity. We confirm that pan-SHIPi is essential by showing that dual treatment with SHIP1 and SHIP2 selective inhibitors reduced adiposity during excess caloric intake. Consistent with this, genetic inactivation of both SHIP paralogs in eosinophils or myeloid cells also reduces obesity and adiposity. In fact, pan-SHIPi requires an eosinophil compartment to prevent diet-induced adiposity, demonstrating that pan-SHIPi acts via an immune mechanism. We also find that pan-SHIPi increases ILC2 cell function in aged, obese mice to reduce their obesity. Finally, we show that pan-SHIPi also reduces hyperglycemia, but not via eosinophils, indicating a separate mechanism for glucose control.
Collapse
Affiliation(s)
- Sandra Fernandes
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Neetu Srivastava
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Chiara Pedicone
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Raki Sudan
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Elizabeth A. Luke
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Otto M. Dungan
- Department of Chemistry, Syracuse University, Syracuse, NY, USA
| | | | - Shea T. Meyer
- Department of Chemistry, Syracuse University, Syracuse, NY, USA
| | - Shawn Dormann
- Department of Chemistry, Syracuse University, Syracuse, NY, USA
| | | | - Benedict J. Chambers
- Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | | | - William G. Kerr
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Chemistry, Syracuse University, Syracuse, NY, USA
- Department of Pediatrics, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
7
|
Wu Y, Yang Z, Cheng K, Bi H, Chen J. Small molecule-based immunomodulators for cancer therapy. Acta Pharm Sin B 2022; 12:4287-4308. [PMID: 36562003 PMCID: PMC9764074 DOI: 10.1016/j.apsb.2022.11.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/31/2022] [Accepted: 11/03/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy has led to a paradigm shift in the treatment of cancer. Current cancer immunotherapies are mostly antibody-based, thus possessing advantages in regard to pharmacodynamics (e.g., specificity and efficacy). However, they have limitations in terms of pharmacokinetics including long half-lives, poor tissue/tumor penetration, and little/no oral bioavailability. In addition, therapeutic antibodies are immunogenic, thus may cause unwanted adverse effects. Therefore, researchers have shifted their efforts towards the development of small molecule-based cancer immunotherapy, as small molecules may overcome the above disadvantages associated with antibodies. Further, small molecule-based immunomodulators and therapeutic antibodies are complementary modalities for cancer treatment, and may be combined to elicit synergistic effects. Recent years have witnessed the rapid development of small molecule-based cancer immunotherapy. In this review, we describe the current progress in small molecule-based immunomodulators (inhibitors/agonists/degraders) for cancer therapy, including those targeting PD-1/PD-L1, chemokine receptors, stimulator of interferon genes (STING), Toll-like receptor (TLR), etc. The tumorigenesis mechanism of various targets and their respective modulators that have entered clinical trials are also summarized.
Collapse
Affiliation(s)
| | | | - Kui Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Huichang Bi
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
8
|
Dungan OM, Dormann S, Fernandes S, Duffy BC, Effiong DG, Kerr WG, Chisholm JD. Synthetic studies on the indane SHIP1 agonist AQX-1125. Org Biomol Chem 2022; 20:4016-4020. [PMID: 35506893 DOI: 10.1039/d2ob00555g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
AQX-1125 is an indane based SHIP1 agonist that has been evaluated in the clinic for the treatment of bladder pain syndrome/interstitial cystitis. To support our own studies on SHIP1 agonists as potential treatments for IBD and Crohn's disease, a new synthetic route to the SHIP1 agonist AQX-1125 has been developed. This sequence utilizes a hydroxy-acid intermediate which allows for ready differentiation of the C6 and C7 positions. The role of the C17 alkene in the biological activity of the system is also investigated, and this functional group is not required for SHIP1 agonist activity. While AQX-1125 shows SHIP1 agonist activity in enzyme assays, it does not show activity in cell based assays similar to other SHIP1 agonists, which limits the utility of this molecule.
Collapse
Affiliation(s)
- Otto M Dungan
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| | - Shawn Dormann
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| | - Sandra Fernandes
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Brian C Duffy
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| | - Daniel G Effiong
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| | - William G Kerr
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA. .,Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.,Department of Pediatrics, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - John D Chisholm
- Department of Chemistry, Syracuse University, 1-014 Center for Science and Technology, Syracuse, NY 13244, USA.
| |
Collapse
|
9
|
Pedicone C, Fernandes S, Matera A, Meyer ST, Loh S, Ha JH, Bernard D, Chisholm JD, Paolicelli RC, Kerr WG. Discovery of a novel SHIP1 agonist that promotes degradation of lipid-laden phagocytic cargo by microglia. iScience 2022; 25:104170. [PMID: 35465359 PMCID: PMC9020084 DOI: 10.1016/j.isci.2022.104170] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/31/2021] [Accepted: 03/24/2022] [Indexed: 12/01/2022] Open
Abstract
Here, we describe the use of artificial intelligence to identify novel agonists of the SH2-containing 5′ inositol phosphatase 1 (SHIP1). One of the compounds, K306, represents the most potent agonist identified to date. We find that K306 exhibits selectivity for SHIP1 vs. the paralog enzyme SHIP2, and this activation does not require the C2 domain of SHIP1 which other known SHIP1 agonists require. Thus, K306 represents a new class of SHIP1 agonists with a novel mode of agonism. Importantly, we find that K306 can suppress induction of inflammatory cytokines and iNOS in macrophages or microglia, but not by their SHIP1-deficient counterparts. K306 also reduces TNF-α production in vivo in an LPS-induced endotoxemia assay. Finally, we show that K306 enhances phagolysosomal degradation of synaptosomes and dead neurons by microglia revealing a novel function for SHIP1 that might be exploited therapeutically in dementia. Discovery of a potent SHIP1 selective agonist (K306) via artificial intelligence SHIP1 agonism via K306 is independent of the C2 domain and increases PI(3,4)P2 levels K306 reduces IL-6, TNF-α, and iNOS induction in microglia and macrophages K306 promotes phagocytic degradation of lipid-laden but not protein cargo in microglia
Collapse
Affiliation(s)
- Chiara Pedicone
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Sandra Fernandes
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Alessandro Matera
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Shea T Meyer
- Chemistry Department, Syracuse University, Syracuse, NY 13210, USA
| | - Stewart Loh
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Jeung-Hoi Ha
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | | | - John D Chisholm
- Chemistry Department, Syracuse University, Syracuse, NY 13210, USA
| | | | - William G Kerr
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.,Chemistry Department, Syracuse University, Syracuse, NY 13210, USA
| |
Collapse
|
10
|
Immune Checkpoint Receptors Signaling in T Cells. Int J Mol Sci 2022; 23:ijms23073529. [PMID: 35408889 PMCID: PMC8999077 DOI: 10.3390/ijms23073529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/16/2022] [Accepted: 03/21/2022] [Indexed: 12/15/2022] Open
Abstract
The characterization of the receptors negatively modulating lymphocyte function is rapidly advancing, driven by success in tumor immunotherapy. As a result, the number of immune checkpoint receptors characterized from a functional perspective and targeted by innovative drugs continues to expand. This review focuses on the less explored area of the signaling mechanisms of these receptors, of those expressed in T cells. Studies conducted mainly on PD-1, CTLA-4, and BTLA have evidenced that the extracellular parts of some of the receptors act as decoy receptors for activating ligands, but in all instances, the tyrosine phosphorylation of their cytoplasmatic tail drives a crucial inhibitory signal. This negative signal is mediated by a few key signal transducers, such as tyrosine phosphatase, inositol phosphatase, and diacylglycerol kinase, which allows them to counteract TCR-mediated activation. The characterization of these signaling pathways is of great interest in the development of therapies for counteracting tumor-infiltrating lymphocyte exhaustion/anergy independently from the receptors involved.
Collapse
|
11
|
Zhao H, Luo F, Xue J, Li S, Xu RH. Emerging immunological strategies: recent advances and future directions. Front Med 2021; 15:805-828. [PMID: 34874513 DOI: 10.1007/s11684-021-0886-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/31/2021] [Indexed: 12/12/2022]
Abstract
Immunotherapy plays a compelling role in cancer treatment and has already made remarkable progress. However, many patients receiving immune checkpoint inhibitors fail to achieve clinical benefits, and the response rates vary among tumor types. New approaches that promote anti-tumor immunity have recently been developed, such as small molecules, bispecific antibodies, chimeric antigen receptor T cell products, and cancer vaccines. Small molecule drugs include agonists and inhibitors that can reach the intracellular or extracellular targets of immune cells participating in innate or adaptive immune pathways. Bispecific antibodies, which bind two different antigens or one antigen with two different epitopes, are of great interest. Chimeric antigen receptor T cell products and cancer vaccines have also been investigated. This review explores the recent progress and challenges of different forms of immunotherapy agents and provides an insight into future immunotherapeutic strategies.
Collapse
Affiliation(s)
- Hongyun Zhao
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Fan Luo
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jinhui Xue
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Su Li
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Rui-Hua Xu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
12
|
So EY, Sun C, Wu KQ, Dubielecka PM, Reginato AM, Liang OD. Inhibition of lipid phosphatase SHIP1 expands myeloid-derived suppressor cells and attenuates rheumatoid arthritis in mice. Am J Physiol Cell Physiol 2021; 321:C569-C584. [PMID: 34288720 DOI: 10.1152/ajpcell.00433.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Rheumatoid arthritis (RA) is a debilitating autoimmune disease of unknown cause, characterized by infiltration and accumulation of activated immune cells in the synovial joints where cartilage and bone destructions occur. Myeloid-derived suppressor cells (MDSCs) are of myeloid origin and are able to suppress T cell responses. Src homology 2 domain containing inositol polyphosphate 5-phosphatase 1 (SHIP1) was shown to be involved in the regulation of MDSC differentiation. The purpose of the present study was to investigate the effect of inhibition of SHIP1 on expansion of MDSCs in RA using a collagen-induced inflammatory arthritis (CIA) mouse model. In DBA/1 mice treatment with a small molecule specific SHIP1 inhibitor 3α-aminocholestane (3AC) induced a marked expansion of MDSCs in vivo. Both pre-treatment with 3AC of DBA/1 mice prior to CIA induction and intervention with 3AC during CIA progression significantly reduced disease incidence and severity. Adoptive transfer of MDSCs isolated from 3AC-treated mice, but not naïve MDSCs from normal mice, into CIA mice significantly reduced disease incidence and severity, indicating that the 3AC-induced MDSCs were the cellular mediators of the observed amelioration of the disease. In conclusion, inhibition of SHIP1 expands MDSCs in vivo and attenuates development of CIA in mice. Small molecule specific inhibition of SHIP1 may therefore offer therapeutic benefit to patients with RA and other autoimmune diseases.
Collapse
Affiliation(s)
- Eui-Young So
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, Rhode Island, United States
| | - Changqi Sun
- Division of Rheumatology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, Rhode Island, United States
| | - Keith Q Wu
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, Rhode Island, United States
| | - Patrycja M Dubielecka
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, Rhode Island, United States
| | - Anthony M Reginato
- Division of Rheumatology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, Rhode Island, United States
| | - Olin D Liang
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, Rhode Island, United States
| |
Collapse
|
13
|
Ecker V, Stumpf M, Brandmeier L, Neumayer T, Pfeuffer L, Engleitner T, Ringshausen I, Nelson N, Jücker M, Wanninger S, Zenz T, Wendtner C, Manske K, Steiger K, Rad R, Müschen M, Ruland J, Buchner M. Targeted PI3K/AKT-hyperactivation induces cell death in chronic lymphocytic leukemia. Nat Commun 2021; 12:3526. [PMID: 34112805 PMCID: PMC8192787 DOI: 10.1038/s41467-021-23752-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/11/2021] [Indexed: 02/05/2023] Open
Abstract
Current therapeutic approaches for chronic lymphocytic leukemia (CLL) focus on the suppression of oncogenic kinase signaling. Here, we test the hypothesis that targeted hyperactivation of the phosphatidylinositol-3-phosphate/AKT (PI3K/AKT)-signaling pathway may be leveraged to trigger CLL cell death. Though counterintuitive, our data show that genetic hyperactivation of PI3K/AKT-signaling or blocking the activity of the inhibitory phosphatase SH2-containing-inositol-5'-phosphatase-1 (SHIP1) induces acute cell death in CLL cells. Our mechanistic studies reveal that increased AKT activity upon inhibition of SHIP1 leads to increased mitochondrial respiration and causes excessive accumulation of reactive oxygen species (ROS), resulting in cell death in CLL with immunogenic features. Our results demonstrate that CLL cells critically depend on mechanisms to fine-tune PI3K/AKT activity, allowing sustained proliferation and survival but avoid ROS-induced cell death and suggest transient SHIP1-inhibition as an unexpectedly promising concept for CLL therapy.
Collapse
MESH Headings
- Animals
- Cell Death/drug effects
- Cell Line, Tumor
- Cell Survival/drug effects
- Disease Progression
- Humans
- Immunohistochemistry
- Leukemia, Lymphocytic, Chronic, B-Cell/enzymology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Mice
- Mice, Transgenic
- Mitochondria/drug effects
- Mitochondria/metabolism
- Oxidative Phosphorylation
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/antagonists & inhibitors
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics
- Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Small Interfering
- RNA-Seq
- Reactive Oxygen Species/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Transplantation, Homologous
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Veronika Ecker
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Martina Stumpf
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Lisa Brandmeier
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Tanja Neumayer
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Lisa Pfeuffer
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Thomas Engleitner
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Ingo Ringshausen
- Wellcome/MRC Cambridge Stem Cell Institute and Department of Haematology, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Nina Nelson
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Wanninger
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
| | - Thorsten Zenz
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Clemens Wendtner
- Munich Clinic Schwabing, Academic Teaching Hospital, Ludwig-Maximilians University (LMU), Munich, Germany
| | - Katrin Manske
- Institute of Molecular Immunology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, Technische Universität München, München, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Roland Rad
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Markus Müschen
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Maike Buchner
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany.
- TranslaTUM - Central Institute for Translational Cancer Research, Technical University of Munich, Munich, Germany.
| |
Collapse
|
14
|
Zhang J, Zhang Y, Qu B, Yang H, Hu S, Dong X. If small molecules immunotherapy comes, can the prime be far behind? Eur J Med Chem 2021; 218:113356. [PMID: 33773287 DOI: 10.1016/j.ejmech.2021.113356] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/15/2021] [Accepted: 02/28/2021] [Indexed: 02/07/2023]
Abstract
Anti-cancer immunotherapy, which includes cellular immunotherapy, immune checkpoint inhibitors and cancer vaccines, has transformed the treatment strategies of several malignancies in the past decades. Immune checkpoints blockade (ICB) is the most commonly tested therapy and has the potential to induce a durable immune response in different types of cancers. However, all approved immune checkpoint inhibitors (ICIs) are monoclonal antibodies (mAbs), which are fraught with disadvantages including lack of oral bioavailability, prolonged tissue retention and poor membrane permeability. Therefore, the research focus has shifted to developing small molecule inhibitors to obviate the limitations of mAbs. Given the complexity of the tumor micro-environment (TME), the combination of ICIs with various small molecule agonists/inhibitors are currently being tested in clinical trials to improve treatment outcomes and prevent tumor recurrence. In this review, we have summarized the mechanisms and therapeutic potential of several molecular targets, along with the current status of small molecule inhibitors.
Collapse
Affiliation(s)
- Jingyu Zhang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Yu Zhang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Bingxue Qu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Haiyan Yang
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), PR China; Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, PR China
| | - Shengquan Hu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China.
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310058, PR China; Cancer Center of Zhejiang University, Hangzhou, 310058, PR China.
| |
Collapse
|
15
|
Targeting SHIP1 and SHIP2 in Cancer. Cancers (Basel) 2021; 13:cancers13040890. [PMID: 33672717 PMCID: PMC7924360 DOI: 10.3390/cancers13040890] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/12/2021] [Accepted: 02/13/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Phosphoinositol signaling pathways and their dysregulation have been shown to have a fundamental role in health and disease, respectively. The SH2-containing 5′ inositol phosphatases, SHIP1 and SHIP2, are regulators of the PI3K/AKT pathway that have crucial roles in cancer progression. This review aims to summarize the role of SHIP1 and SHIP2 in cancer signaling and the immune response to cancer, the discovery and use of SHIP inhibitors and agonists as possible cancer therapeutics. Abstract Membrane-anchored and soluble inositol phospholipid species are critical mediators of intracellular cell signaling cascades. Alterations in their normal production or degradation are implicated in the pathology of a number of disorders including cancer and pro-inflammatory conditions. The SH2-containing 5′ inositol phosphatases, SHIP1 and SHIP2, play a fundamental role in these processes by depleting PI(3,4,5)P3, but also by producing PI(3,4)P2 at the inner leaflet of the plasma membrane. With the intent of targeting SHIP1 or SHIP2 selectively, or both paralogs simultaneously, small molecule inhibitors and agonists have been developed and tested in vitro and in vivo over the last decade in various disease models. These studies have shown promising results in various pre-clinical models of disease including cancer and tumor immunotherapy. In this review the potential use of SHIP inhibitors in cancer is discussed with particular attention to the molecular structure, binding site and efficacy of these SHIP inhibitors.
Collapse
|
16
|
Sugawara S, Manickam C, Reeves K. TRIGGERED: could refocused cell signaling be key to natural killer cell-based HIV immunotherapeutics? AIDS 2021; 35:165-176. [PMID: 33116071 PMCID: PMC7775286 DOI: 10.1097/qad.0000000000002743] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Natural killer (NK) cells are one of the critical innate immune effector cells that directly kill tumors and virus-infected cells, and modulate other immune cells including dendritic cells, CD4+ and CD8+ T cells. Signals from activating and inhibitory surface receptors orchestrate the regulatory and cytotoxic functions of NK cells. Although a number of surface receptors are involved, multiple signaling molecules are shared so that NK cell responses are synergistically regulated. Many pathogens and tumors evade NK cell responses by targeting NK cell signaling. Particularly in HIV/simian immunodeficiency virus (SIV) infection, the NK cell repertoire is diminished by changes in subsets of NK cells, expression of activating and inhibitory receptors, and intracellular signaling molecules. However, in-depth studies on intracellular signaling in NK cells in HIV/SIV infections remain limited. Checkpoint blockade and chimeric antigen receptor (CAR)-NK cells have demonstrated enhanced NK cell activities against tumors and viral infections. In addition, targeting intracellular signaling molecules by small molecules could also improve NK cell responses towards HIV/SIV infection in vivo. Therefore, further understanding of NK cell signaling including identification of key signaling molecules is crucial to maximize the efficacy of NK cell-based treatments. Herein, we review the current state of the literature and outline potential future avenues where optimized NK cells could be utilized in HIV-1 cure strategies and other immunotherapeutics in PLWH.
Collapse
Affiliation(s)
- Sho Sugawara
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Cordelia Manickam
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Ragon Institute of Massachusetts General Hospital, MIT, and Harvard, Cambridge, MA
| |
Collapse
|
17
|
UV Light-inactivated HSV-1 Stimulates Natural Killer Cell-induced Killing of Prostate Cancer Cells. J Immunother 2020; 42:162-174. [PMID: 30933043 DOI: 10.1097/cji.0000000000000261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Herein we demonstrate that ultraviolet light-inactivated Herpes Simplex Virus-1 (UV-HSV-1) stimulates peripheral blood mononuclear cells (PBMCs) to lyse both androgen-sensitive and androgen-independent prostate cancer (PrCA) cell lines, but not the benign prostatic hyperplastic epithelial cell line, BPH-1, and is 1000-10,000-fold more potent at stimulating this killing than ultraviolet light-inactivated Vesicular Stomatitis Virus, adenovirus, reovirus or cytomegalovirus. Among PBMCs, natural killer (NK) cells appear to be a major cell type involved in this killing and UV-HSV-1 appears to directly and potently stimulate NK cell expression of CD69, degranulation, cytokine production, and migration to IL-8 in PC3 conditioned medium. We also found that UV-HSV-1 stimulates glycolysis in PBMCs and NK cells, and that 2-deoxyglucose and the protein kinase C inhibitor, Go6976, and the NFκB inhibitor, Bay 11-7082, all abrogate UV-HSV-1 activated killing of PC3 cells by PBMCs and NK cells. Using neutralizing anti-Toll-like receptor 2 (TLR2) we found that UV-HSV-1, like HSV-1, activates NK cells via TLR2. Taken together, these results are consistent with Toll-like receptor 2 ligands on UV-HSV-1 stimulating TLR2 on NK cells to activate protein kinase C, leading to enhanced glycolysis and NFκB activation, both of which play a critical role in this anti-PrCA innate immune response. Importantly, UV-HSV-1 synergizes with IL-15 to increase the cytolytic activity of PBMCs against PC3 cells and there was considerable donor-to-donor variation in killing ability. These results support the preclinical development of UV-HSV-1 as an adjuvant, in combination with IL-15, for cell infusions of healthy, preselected NK cells to treat PrCA.
Collapse
|
18
|
Clement D, Goodridge JP, Grimm C, Patel S, Malmberg KJ. TRP Channels as Interior Designers: Remodeling the Endolysosomal Compartment in Natural Killer Cells. Front Immunol 2020; 11:753. [PMID: 32411146 PMCID: PMC7198808 DOI: 10.3389/fimmu.2020.00753] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/02/2020] [Indexed: 12/15/2022] Open
Abstract
Cytotoxic lymphocytes, including natural killer (NK) cells and T cells are distinguished by their ability to eliminate target cells through release of secretory lysosomes. Conventional lysosomes and secretory lysosomes are part of the pleomorphic endolysosomal system and characterized by its highly dynamic nature. Several calcium-permeable TRP calcium channels play an essential role in endolysosomal calcium signaling to ensure proper function of these organelles. In NK cells, the expression of self MHC-specific inhibitory receptors dynamically tunes their secretory potential in a non-transcriptional, calcium-dependent manner. New insights suggest that TRPML1-mediated lysosomal calcium fluxes are tightly interconnected to NK cell functionality through modulation of granzyme B and perforin content of the secretory lysosome. Lysosomal TRP channels show a subset-specific expression pattern during NK differentiation, which is paralleled with gradually increased loading of effector molecules in secretory lysosomes. Methodological advances, including organellar patch-clamping, specific pharmacological modulators, and genetically-encoded calcium indicators open up new possibilities to investigate how TRP channels influence communication between intracellular organelles in immune cells. This review discusses our current understanding of lysosome biogenesis in NK cells with an emphasis on the TRP mucolipin family and the implications for NK cell functionality and cancer immunotherapy.
Collapse
Affiliation(s)
- Dennis Clement
- The KG Jebsen Center for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cancer Immunology, Oslo University Hospital, Institute for Cancer Research, Oslo, Norway
| | | | - Christian Grimm
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Karl-Johan Malmberg
- The KG Jebsen Center for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cancer Immunology, Oslo University Hospital, Institute for Cancer Research, Oslo, Norway
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
19
|
Small molecule targeting of SHIP1 and SHIP2. Biochem Soc Trans 2020; 48:291-300. [DOI: 10.1042/bst20190775] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
Abstract
Modulating the activity of the Src Homology 2 (SH2) — containing Inositol 5′-Phosphatase (SHIP) enzyme family with small molecule inhibitors provides a useful and unconventional method of influencing cell signaling in the PI3K pathway. The development of small molecules that selectively target one of the SHIP paralogs (SHIP1 or SHIP2) as well as inhibitors that simultaneously target both enzymes have provided promising data linking the phosphatase activity of the SHIP enzymes to disorders and disease states that are in dire need of new therapeutic targets. These include cancer, immunotherapy, diabetes, obesity, and Alzheimer's disease. In this mini-review, we will provide a brief overview of research in these areas that support targeting SHIP1, SHIP2 or both enzymes for therapeutic purposes.
Collapse
|
20
|
Pedicone C, Fernandes S, Dungan OM, Dormann SM, Viernes DR, Adhikari AA, Choi LB, De Jong EP, Chisholm JD, Kerr WG. Pan-SHIP1/2 inhibitors promote microglia effector functions essential for CNS homeostasis. J Cell Sci 2020; 133:jcs238030. [PMID: 31780579 PMCID: PMC10682645 DOI: 10.1242/jcs.238030] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
We show here that both SHIP1 (Inpp5d) and its paralog SHIP2 (Inppl1) are expressed at protein level in microglia. To examine whether targeting of SHIP paralogs might influence microglial physiology and function, we tested the capacity of SHIP1-selective, SHIP2-selective and pan-SHIP1/2 inhibitors for their ability to impact on microglia proliferation, lysosomal compartment size and phagocytic function. We find that highly potent pan-SHIP1/2 inhibitors can significantly increase lysosomal compartment size, and phagocytosis of dead neurons and amyloid beta (Aβ)1-42 by microglia in vitro We show that one of the more-potent and water-soluble pan-SHIP1/2 inhibitors, K161, can penetrate the blood-brain barrier. Consistent with this, K161 increases the capacity of CNS-resident microglia to phagocytose Aβ and apoptotic neurons following systemic administration. These findings provide the first demonstration that small molecule modulation of microglia function in vivo is feasible, and suggest that dual inhibition of the SHIP1 and 2 paralogs can provide a novel means to enhance basal microglial homeostatic functions for therapeutic purposes in Alzheimer's disease and, possibly, other types of dementia where increased microglial function could be beneficial.
Collapse
Affiliation(s)
- Chiara Pedicone
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Sandra Fernandes
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Otto M Dungan
- Department of Chemistry, Syracuse University, Syracuse, NY 13210, USA
| | - Shawn M Dormann
- Department of Chemistry, Syracuse University, Syracuse, NY 13210, USA
| | - Dennis R Viernes
- Department of Chemistry, Syracuse University, Syracuse, NY 13210, USA
| | - Arijit A Adhikari
- Department of Chemistry, Syracuse University, Syracuse, NY 13210, USA
| | - Lydia B Choi
- Department of Chemistry, Syracuse University, Syracuse, NY 13210, USA
| | - Ebbing P De Jong
- Proteomics and Mass Spectrometry Core Facility, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - John D Chisholm
- Department of Chemistry, Syracuse University, Syracuse, NY 13210, USA
| | - William G Kerr
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Chemistry, Syracuse University, Syracuse, NY 13210, USA
- Department of Pediatrics, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
21
|
Pal Singh S, de Bruijn MJW, Velaso Gago da Graça C, Corneth OBJ, Rip J, Stadhouders R, Meijers RWJ, Schurmans S, Kerr WG, Ter Burg J, Eldering E, Langerak AW, Pillai SY, Hendriks RW. Overexpression of SH2-Containing Inositol Phosphatase Contributes to Chronic Lymphocytic Leukemia Survival. THE JOURNAL OF IMMUNOLOGY 2019; 204:360-374. [PMID: 31836657 DOI: 10.4049/jimmunol.1900153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 11/11/2019] [Indexed: 01/01/2023]
Abstract
Balanced activity of kinases and phosphatases downstream of the BCR is essential for B cell differentiation and function and is disturbed in chronic lymphocytic leukemia (CLL). In this study, we employed IgH.TEμ mice, which spontaneously develop CLL, and stable EMC CLL cell lines derived from these mice to explore the role of phosphatases in CLL. Genome-wide expression profiling comparing IgH.TEμ CLL cells with wild-type splenic B cells identified 96 differentially expressed phosphatase genes, including SH2-containing inositol phosphatase (Ship2). We found that B cell-specific deletion of Ship2, but not of its close homolog Ship1, significantly reduced CLL formation in IgH.TEμ mice. Treatment of EMC cell lines with Ship1/2 small molecule inhibitors resulted in the induction of caspase-dependent apoptosis. Using flow cytometry and Western blot analysis, we observed that blocking Ship1/2 abrogated EMC cell survival by exerting dual effects on the BCR signaling cascade. On one hand, specific Ship1 inhibition enhanced calcium signaling and thereby abrogated an anergic response to BCR stimulation in CLL cells. On the other hand, concomitant Ship1/Ship2 inhibition or specific Ship2 inhibition reduced constitutive activation of the mTORC1/ribosomal protein S6 pathway and downregulated constitutive expression of the antiapoptotic protein Mcl-1, in both EMC cell lines and primary IgH.TEμ CLL cells. Importantly, also in human CLL, we found overexpression of many phosphatases including SHIP2. Inhibition of SHIP1/SHIP2 reduced cellular survival and S6 phosphorylation and enhanced basal calcium levels in human CLL cells. Taken together, we provide evidence that SHIP2 contributes to CLL pathogenesis in mouse and human CLL.
Collapse
Affiliation(s)
- Simar Pal Singh
- Department of Pulmonary Medicine, Erasmus MC, NL 3000 CA Rotterdam, the Netherlands.,Department of Immunology, Erasmus MC, NL 3000 CA Rotterdam, the Netherlands.,Postgraduate School Molecular Medicine, Erasmus MC, NL 3000 CA Rotterdam, the Netherlands
| | | | | | - Odilia B J Corneth
- Department of Pulmonary Medicine, Erasmus MC, NL 3000 CA Rotterdam, the Netherlands
| | - Jasper Rip
- Department of Pulmonary Medicine, Erasmus MC, NL 3000 CA Rotterdam, the Netherlands
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus MC, NL 3000 CA Rotterdam, the Netherlands.,Department of Cell Biology, Erasmus MC, NL 3000 CA Rotterdam, the Netherlands
| | - Ruud W J Meijers
- Department of Immunology, Erasmus MC, NL 3000 CA Rotterdam, the Netherlands
| | - Stéphane Schurmans
- Laboratoire de Génétique Fonctionnelle, GIGA-Research Centre, Université de Liège, 4000 Liège, Belgium
| | - William G Kerr
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210; and
| | - Johanna Ter Burg
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Eric Eldering
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Anton W Langerak
- Department of Immunology, Erasmus MC, NL 3000 CA Rotterdam, the Netherlands
| | - Saravanan Y Pillai
- Department of Pulmonary Medicine, Erasmus MC, NL 3000 CA Rotterdam, the Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, NL 3000 CA Rotterdam, the Netherlands;
| |
Collapse
|
22
|
Saz-Leal P, Del Fresno C, Brandi P, Martínez-Cano S, Dungan OM, Chisholm JD, Kerr WG, Sancho D. Targeting SHIP-1 in Myeloid Cells Enhances Trained Immunity and Boosts Response to Infection. Cell Rep 2019; 25:1118-1126. [PMID: 30380404 PMCID: PMC6226423 DOI: 10.1016/j.celrep.2018.09.092] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/14/2018] [Accepted: 09/27/2018] [Indexed: 12/18/2022] Open
Abstract
β-Glucan-induced trained immunity in myeloid cells leads to long-term protection against secondary infections. Although previous studies have characterized this phenomenon, strategies to boost trained immunity remain undefined. We found that β-glucan-trained macrophages from mice with a myeloid-specific deletion of the phosphatase SHIP-1 (LysMΔSHIP-1) showed enhanced proinflammatory cytokine production in response to lipopolysaccharide. Following β-glucan training, SHIP-1-deficient macrophages exhibited increased phosphorylation of Akt and mTOR targets, correlating with augmented glycolytic metabolism. Enhanced training in the absence of SHIP-1 relied on histone methylation and acetylation. Trained LysMΔSHIP-1 mice produced increased amounts of proinflammatory cytokines upon rechallenge in vivo and were better protected against Candida albicans infection compared with control littermates. Pharmacological inhibition of SHIP-1 enhanced trained immunity against Candida infection in mouse macrophages and human peripheral blood mononuclear cells. Our data establish proof of concept for improvement of trained immunity and a strategy to achieve it by targeting SHIP-1.
Collapse
Affiliation(s)
- Paula Saz-Leal
- Immunobiology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Carlos Del Fresno
- Immunobiology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Paola Brandi
- Immunobiology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Sarai Martínez-Cano
- Immunobiology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Otto M Dungan
- Department of Chemistry, Syracuse University, Syracuse, NY 13210, USA
| | - John D Chisholm
- Department of Chemistry, Syracuse University, Syracuse, NY 13210, USA
| | - William G Kerr
- Department of Chemistry, Syracuse University, Syracuse, NY 13210, USA; Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY 13210, USA; Pediatrics Department, SUNY Upstate Medical University, Syracuse, NY, USA; Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - David Sancho
- Immunobiology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, Madrid, 28029, Spain.
| |
Collapse
|
23
|
Kerr WG, Chisholm JD. The Next Generation of Immunotherapy for Cancer: Small Molecules Could Make Big Waves. THE JOURNAL OF IMMUNOLOGY 2019; 202:11-19. [PMID: 30587569 DOI: 10.4049/jimmunol.1800991] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/20/2018] [Indexed: 12/30/2022]
Abstract
After decades of intense effort, therapeutics that leverage the immune system to fight cancer have now been conclusively demonstrated to be effective. Immuno-oncology has arrived and will play a key role in the treatment of cancer for the foreseeable future. However, the search for novel methods to improve immune responses to cancer continues unabated. Toward this end, small molecules that can either reduce immune suppression in the tumor milieu or enhance activation of cytotoxic lymphocyte responses to the tumor are actively being pursued. Such novel treatment strategies might be used as monotherapies or combined with other cancer therapies to increase and broaden their efficacy. In this article, we provide an overview of small molecule immunotherapeutic approaches for the treatment of cancer. Over the next decade and beyond, these approaches could further enhance our ability to harness the immune system to combat cancer and thus become additional weapons in the oncologist's armory.
Collapse
Affiliation(s)
- William G Kerr
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210; .,Department of Chemistry, Syracuse University, Syracuse, NY 13244; and.,Department of Pediatrics, SUNY Upstate Medical University, Syracuse, NY 13210
| | - John D Chisholm
- Department of Chemistry, Syracuse University, Syracuse, NY 13244; and
| |
Collapse
|
24
|
Ramos AR, Ghosh S, Dedobbeleer M, Robe PA, Rogister B, Erneux C. Lipid phosphatases SKIP and SHIP2 regulate fibronectin-dependent cell migration in glioblastoma. FEBS J 2019; 286:1120-1135. [PMID: 30695232 DOI: 10.1111/febs.14769] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 11/08/2018] [Accepted: 01/25/2019] [Indexed: 12/19/2022]
Abstract
Cell migration is an important process that occurs during development and has also been linked to the motility of cancer cells. Cytoskeleton reorganization takes place in the migration process leading to lamellipodia formation. Understanding the molecular underpinnings of cell migration is particularly important in studies of glioblastoma, a highly invasive and aggressive cancer type. Two members of the phosphoinositide 5-phosphatase family, SKIP and SHIP2, have been associated with cell migration in glioblastoma; however, the precise role these enzymes play in the process-and whether they work in concert-remains unclear. Here, we compared phosphoinositide 5-phosphatases expression in glioblastoma primary cells and cell lines and showed that SHIP2 and SKIP expression greatly varies between different cell types, while OCRL, another phosphoinositide 5-phosphatase, is constitutively expressed. Upon adhesion of U-251 MG cells to fibronectin, SHIP2, SKIP, and PI(4,5)P2 colocalized in membrane ruffles. Upregulation of PI(4,5)P2 was observed in SKIP-depleted U-251 MG cells compared to control cells, but only when cells were adhered to fibronectin. Both PTEN-deficient (U-251) and PTEN-containing (LN229) glioblastoma cells showed a decrease in cell migration velocity in response to SKIP downregulation. Moreover, a SHIP2 catalytic inhibitor lowered cell migration velocity in the U-251 MG cell line. We conclude that integrin activation in U-251 cells leads to colocalization of both SKIP and SHIP2 in ruffles, where they act as potential drivers of cell migration. Depending on their expression levels in glioblastoma, phosphoinositide 5-phosphatases could cooperate and synergize in the regulation of cell migration and adhesion.
Collapse
Affiliation(s)
| | | | | | - Pierre A Robe
- Department of Neurology and Neurosurgery, Utrecht University Medical Center, The Netherlands
| | - Bernard Rogister
- GIGA-Neurosciences Research Center, Université de Liège, Belgium
| | | |
Collapse
|
25
|
Lucca LE, Axisa PP, Singer ER, Nolan NM, Dominguez-Villar M, Hafler DA. TIGIT signaling restores suppressor function of Th1 Tregs. JCI Insight 2019; 4:124427. [PMID: 30728325 DOI: 10.1172/jci.insight.124427] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/03/2019] [Indexed: 12/28/2022] Open
Abstract
Th1 Tregs are characterized by the acquisition of proinflammatory cytokine secretion and reduced suppressor activity. Th1 Tregs are found at increased frequency in autoimmune diseases, including type 1 diabetes and multiple sclerosis (MS). We have previously reported that in vitro stimulation with IL-12 recapitulates the functional and molecular features of MS-associated Th1 Tregs, revealing a central role for hyperactivation of the Akt pathway in their induction. TIGIT is a newly identified coinhibitory receptor that marks Tregs that specifically control Th1 and Th17 responses. Here, we report that signaling through TIGIT counteracts the action of IL-12 in inducing the Th1 program. Specifically, TIGIT signaling represses production of IFN-γ and T-bet expression and restores suppressor function in Tregs treated with IL-12. FoxO1 functional inhibition abolishes the protective effect of TIGIT, indicating that TIGIT signaling promotes FoxO1 nuclear localization. Consistent with this observation, signaling through TIGIT leads to a rapid suppression of Akt function and FoxO1 phosphorylation. Finally, TIGIT stimulation reduces the production of IFN-γ and corrects the suppressor defect of Tregs from patients with MS. Our results indicate an important role for TIGIT in controlling the functional stability of Tregs through repression of Akt, suggesting that the TIGIT pathway could be targeted for immunomodulatory therapies in human autoimmune disorders.
Collapse
Affiliation(s)
- Liliana E Lucca
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Pierre-Paul Axisa
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Emily R Singer
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Neal M Nolan
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - David A Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
26
|
So EY, Sun C, Reginato AM, Dubielecka PM, Ouchi T, Liang OD. Loss of lipid phosphatase SHIP1 promotes macrophage differentiation through suppression of dendritic cell differentiation. Cancer Biol Ther 2018; 20:201-211. [PMID: 30277839 DOI: 10.1080/15384047.2018.1523846] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
SH2-containing inositol 5'-phosphatase-1 (SHIP1) deficiency in mice results in abnormal myeloid expansion, and proinflammatory conditions in the lung. However, the mechanisms involved in SHIP1-mediated regulation of myeloid differentiation remain unclear. Here we show that SHIP1 is a key regulator of early differentiation for dendritic cells (DCs). We also provide critical evidence to modify the function of SHIP1 in in vitro development of BMDCs using the recent framework of defining DCs. We found that loss of SHIP1 suppresses GM-CSF-induced formation of bone marrow-derived DC (BMDC) colonies, leading to reduced BMDC number in BM cell culture. The number of maturated BMDCs decreased in SHIP1-KO culture, due to reduction of immature BMDCs, suggesting SHIP1 is critical for lineage commitment rather than for maturation from myeloid precursors to DCs. We further showed that F4/80+/MHCIIlow BM macrophage-like cells (BMMs) were the main population of SHIP1-KO BM culture. Treatment of wild-type BM culture with 3 α-aminocholestane (3AC), a specific inhibitor for functional activity of SHIP1, caused a similar developmental defect in BMDCs as seen in SHIP1-KO cells, resulting in the absence of BMDC colony, and increased number of BMMs in BM culture. In conclusion, our results suggest that differentiation of BMDCs are markedly impaired under SHIP1 deficient condition, which causes skewed development of myeloid lineage cells manifested as pathological conditions associated with an excess of macrophage population.
Collapse
Affiliation(s)
- Eui Young So
- a Division of Hematology and Oncology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| | - Changqi Sun
- b Division of Rheumatology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| | - Anthony M Reginato
- b Division of Rheumatology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| | - Patrycia M Dubielecka
- a Division of Hematology and Oncology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| | - Toru Ouchi
- c Department of Cancer Genetics , Roswell Park Cancer Institute , Buffalo , USA
| | - Olin D Liang
- a Division of Hematology and Oncology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| |
Collapse
|
27
|
Ramos AR, Ghosh S, Erneux C. The impact of phosphoinositide 5-phosphatases on phosphoinositides in cell function and human disease. J Lipid Res 2018; 60:276-286. [PMID: 30194087 DOI: 10.1194/jlr.r087908] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/01/2018] [Indexed: 02/06/2023] Open
Abstract
Phosphoinositides (PIs) are recognized as major signaling molecules in many different functions of eukaryotic cells. PIs can be dephosphorylated by multiple phosphatase activities at the 5-, 4-, and 3- positions. Human PI 5-phosphatases belong to a family of 10 members. Except for inositol polyphosphate 5-phosphatase A, they all catalyze the dephosphorylation of PI(4,5)P2 and/or PI(3,4,5)P3 at the 5- position. PI 5-phosphatases thus directly control the levels of PI(3,4,5)P3 and participate in the fine-tuning regulatory mechanisms of PI(3,4)P2 and PI(4,5)P2 Second messenger functions have been demonstrated for PI(3,4)P2 in invadopodium maturation and lamellipodia formation. PI 5-phosphatases can use several substrates on isolated enzymes, and it has been challenging to establish their real substrate in vivo. PI(4,5)P2 has multiple functions in signaling, including interacting with scaffold proteins, ion channels, and cytoskeleton proteins. PI 5-phosphatase isoenzymes have been individually implicated in human diseases, such as the oculocerebrorenal syndrome of Lowe, through mechanisms that include lipid control. Oncogenic and tumor-suppressive functions of PI 5-phosphatases have also been reported in different cell contexts. The mechanisms responsible for genetic diseases and for oncogenic or tumor-suppressive functions are not fully understood. The regulation of PI 5-phosphatases is thus crucial in understanding cell functions.
Collapse
Affiliation(s)
- Ana Raquel Ramos
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Somadri Ghosh
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Christophe Erneux
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, 1070 Brussels, Belgium
| |
Collapse
|
28
|
Blaya D, Aguilar-Bravo B, Hao F, Casacuberta-Serra S, Coll M, Perea L, Vallverdú J, Graupera I, Pose E, Llovet L, Barquinero J, Cubero FJ, Caballería J, Ginès P, Sancho-Bru P. Expression of microRNA-155 in inflammatory cells modulates liver injury. Hepatology 2018; 68:691-706. [PMID: 29420849 PMCID: PMC6082738 DOI: 10.1002/hep.29833] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 12/17/2022]
Abstract
UNLABELLED MicroRNA 155 (miR-155) is involved in immune and inflammatory diseases and is associated with liver fibrosis and steatohepatitis. However, the mechanisms involved in miR-155 regulation of liver injury are largely unknown. The role of miR-155 in acute liver injury was assessed in wild-type (WT), miR-155-/- , and miR-155-/- mice transplanted with WT bone marrow. Additionally, miR-155 expression was evaluated in liver tissue and peripheral blood mononuclear cells of patients with autoimmune hepatitis. Concanavalin A, but not acetaminophen, treatment increased the expression of miR-155 in liver tissue of WT mice. Concanavalin A induced increases in cell death, liver aminotransferases, and expression of proinflammatory cytokines (chemokine [C-X-C motif] ligands 1, 5, 9, 10, and 11; chemokine [C-C motif] ligands 2 and 20; and intercellular cell adhesion molecule 1) in miR-155-/- compared to WT mice. Importantly, these animals showed a significant decrease in cluster of differentiation 4-positive/chemokine (C-X-C motif) receptor 3-positive and forkhead box p3-positive cell recruitment but no changes in other inflammatory cell populations. Mechanistically, miR-155-deficient regulatory T cells showed increased SH2 domain-containing inositol 5-phosphatase 1 expression, a known target of miR-155. Inhibition of SH2 domain-containing inositol 5-phosphatase 1 in miR-155-/- mice restored forkhead box p3 recruitment and reduced liver cytokine expression. Transplantation of bone marrow from WT animals into miR-155-/- mice partially reversed the effect of concanavalin A on miR-155-/- mice as assessed by proinflammatory cytokines and cell death protein expression. Patients with autoimmune hepatitis showed a marked increase in miR-155 expression in the liver but reduced expression of miR-155 in peripheral blood mononuclear cells. CONCLUSION miR-155 expression is altered in both liver tissue and circulating inflammatory cells during liver injury, thus regulating inflammatory cell recruitment and liver damage; these results suggest that maintaining miR-155 expression in inflammatory cells might be a potential strategy to modulate liver injury. (Hepatology 2018).
Collapse
Affiliation(s)
- Delia Blaya
- Laboratory of Liver Cell Plasticity and Tissue Repair, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Beatriz Aguilar-Bravo
- Laboratory of Liver Cell Plasticity and Tissue Repair, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Fengjie Hao
- Department of Immunology, Complutense University School of Medicine, Madrid, Spain,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | | | - Mar Coll
- Laboratory of Liver Cell Plasticity and Tissue Repair, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Luis Perea
- Laboratory of Liver Cell Plasticity and Tissue Repair, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Júlia Vallverdú
- Laboratory of Liver Cell Plasticity and Tissue Repair, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Isabel Graupera
- Laboratory of Liver Cell Plasticity and Tissue Repair, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain,Liver Unit, Hospital Clínic, Barcelona, Spain
| | - Elisa Pose
- Laboratory of Liver Cell Plasticity and Tissue Repair, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain,Liver Unit, Hospital Clínic, Barcelona, Spain
| | - Laura Llovet
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain,Liver Unit, Hospital Clínic, Barcelona, Spain
| | - Jordi Barquinero
- Gene and Cell Therapy Laboratory, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Complutense University School of Medicine, Madrid, Spain,12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Juan Caballería
- Laboratory of Liver Cell Plasticity and Tissue Repair, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain,Liver Unit, Hospital Clínic, Barcelona, Spain
| | - Pere Ginès
- Laboratory of Liver Cell Plasticity and Tissue Repair, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain,Liver Unit, Hospital Clínic, Barcelona, Spain
| | - Pau Sancho-Bru
- Laboratory of Liver Cell Plasticity and Tissue Repair, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| |
Collapse
|
29
|
Zhou M, Dai J, Zhou Y, Wu J, Xu T, Zhou D, Wang X. Propofol improves the function of natural killer cells from the peripheral blood of patients with esophageal squamous cell carcinoma. Exp Ther Med 2018; 16:83-92. [PMID: 29977357 PMCID: PMC6030861 DOI: 10.3892/etm.2018.6140] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/05/2018] [Indexed: 12/16/2022] Open
Abstract
Postoperative immunosuppression is associated with the recurrence and metastasis of esophageal squamous cell carcinoma (ESCC). Propofol is a commonly used intravenous anesthetic and has been reported to be associated with immunosuppression; however, little is known about its effect on innate immune cells during the postoperative period in patients with ESCC. The aim of the present study was to investigate the effects of propofol on the phenotype and cytotoxicity of natural killer (NK) cells derived from the peripheral blood of patients with ESCC. The percentage, phenotype and function of NK cells were compared between patients with ESCC and healthy volunteers using flow cytometry. NK cells were negatively sorted using magnetic beads and cocultured with propofol to assess changes in phenotype and function. The results revealed that the percentage of NK cells was significantly increased in the peripheral blood of patients with ESCC, while their activity and cytotoxicity were impaired. NK cells were successfully separated from peripheral blood in vitro and it was demonstrated that propofol enhanced their activity by influencing the expression of activating or inhibitory receptors. Furthermore, propofol was able to increase the cytotoxicity of NK cells from the peripheral blood of patients with ESCC. These results suggest that propofol is able to improve the function of NK cells in patients with ESCC and may therefore be an appropriate anesthetic for ESCC surgery.
Collapse
Affiliation(s)
- Min Zhou
- Department of Anesthesiology, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Junchao Dai
- Department of Anesthesiology, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yu Zhou
- Department of Anesthesiology, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jian Wu
- Department of Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Tao Xu
- Department of Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Denglian Zhou
- Dean's Office, South West Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xiaobin Wang
- Department of Anesthesiology, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
30
|
Abstract
Future immunotherapies may stimulate immune system attack as well as memory against cancer to prevent relapse in patients.
Collapse
|
31
|
Ghosh S, Scozzaro S, Ramos AR, Delcambre S, Chevalier C, Krejci P, Erneux C. Inhibition of SHIP2 activity inhibits cell migration and could prevent metastasis in breast cancer cells. J Cell Sci 2018; 131:jcs.216408. [DOI: 10.1242/jcs.216408] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 07/06/2018] [Indexed: 12/13/2022] Open
Abstract
Metastasis of breast cancer cells to distant organs is responsible for approximately 50 % in cancer related deaths in women worldwide. SHIP2 is a phosphoinositide 5-phosphatase for PI(3,4,5)P3 and PI(4,5)P2. Through depletion of SHIP2 in triple negative MDA-MB-231 cells and the use of SHIP2 inhibitors, it appeared that cell migration is positively controlled by SHIP2. The effect of SHIP2 on migration, observed in MDA-MB-231 cells, appears to be mediated by PI(3,4)P2. Adhesion on fibronectin is always increased in SHIP2 depleted cells. Apoptosis measured in MDA-MB-231 cells is also increased in SHIP2 depleted cells as compared to control cells. In xenograft mice, SHIP2 depleted MDA-MB-231 cells form significantly smaller tumors compared to control cells and less metastasis detected in lung sections. Our data reveal a general role of SHIP2 in the control of cell migration in breast cancer cells and a second messenger role for PI(3,4)P2 in the migration mechanism. In this model, SHIP2 function on apoptosis on cells incubated in vitro, or in mice tumor digested cells, could account for its role on tumor growth determined in vivo.
Collapse
Affiliation(s)
- Somadri Ghosh
- IRIBHM, Campus Erasme, ULB Bâtiment C, 808 route de Lennik 1070 Bruxelles, Belgium
| | - Samuel Scozzaro
- IRIBHM, Campus Erasme, ULB Bâtiment C, 808 route de Lennik 1070 Bruxelles, Belgium
| | - Ana Raquel Ramos
- IRIBHM, Campus Erasme, ULB Bâtiment C, 808 route de Lennik 1070 Bruxelles, Belgium
| | | | - Clément Chevalier
- Center for Microscopy and Molecular Imaging ULB, 12 rue des professeurs Jeener et Brachet, 6041 Charleroi, Belgium
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic
| | - Christophe Erneux
- IRIBHM, Campus Erasme, ULB Bâtiment C, 808 route de Lennik 1070 Bruxelles, Belgium
| |
Collapse
|
32
|
Liu CF, Min XY, Wang N, Wang JX, Ma N, Dong X, Zhang B, Wu W, Li ZF, Zhou W, Li K. Complement Receptor 3 Has Negative Impact on Tumor Surveillance through Suppression of Natural Killer Cell Function. Front Immunol 2017; 8:1602. [PMID: 29209332 PMCID: PMC5702005 DOI: 10.3389/fimmu.2017.01602] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/06/2017] [Indexed: 01/31/2023] Open
Abstract
Complement receptor 3 (CR3) is expressed abundantly on natural killer (NK) cells; however, whether it plays roles in NK cell-dependent tumor surveillance is largely unknown. Here, we show that CR3 is an important negative regulator of NK cell function, which has negative impact on tumor surveillance. Mice deficient in CR3 (CD11b-/- mice) exhibited a more activated NK phenotype and had enhanced NK-dependent tumor killing. In a B16-luc melanoma-induced lung tumor growth and metastasis model, mice deficient in CR3 had reduced tumor growth and metastases, compared with WT mice. In addition, adaptive transfer of NK cells lacking CR3 (into NK-deficient mice) mediated more efficient suppression of tumor growth and metastases, compared with the transfer of CR3 sufficient NK cells, suggesting that CR3 can impair tumor surveillance through suppression of NK cell function. In vitro analyses showed that engagement of CR3 with iC3b (classical CR3 ligand) on NK cells negatively regulated NK cell activity and effector functions (i.e. direct tumor cell killing, antibody-dependent NK-mediated tumor killing). Cell signaling analyses showed that iC3b stimulation caused activation of Src homology 2 domain-containing inositol-5-phosphatase-1 (SHIP-1) and JNK, and suppression of ERK in NK cells, supporting that iC3b mediates negative regulation of NK cell function through its effects on SHIP-1, JNK, and ERK signal transduction pathways. Thus, our findings demonstrate a previously unknown role for CR3 in dysregulation of NK-dependent tumor surveillance and suggest that the iC3b/CR3 signaling is a critical negative regulator of NK cell function and may represent a new target for preserving NK cell function in cancer patients and improving NK cell-based therapy.
Collapse
Affiliation(s)
- Cheng-Fei Liu
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Xiao-Yun Min
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Naiyin Wang
- Medical Research Council (MRC) Centre for Transplantation, King's College London, Guy's Hospital, London, United Kingdom
| | - Jia-Xing Wang
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Ning Ma
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Xia Dong
- Medical Research Council (MRC) Centre for Transplantation, King's College London, Guy's Hospital, London, United Kingdom
| | - Bing Zhang
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Weiju Wu
- Medical Research Council (MRC) Centre for Transplantation, King's College London, Guy's Hospital, London, United Kingdom
| | - Zong-Fang Li
- National Local Joint Engineering Research Centre of Biodiagnostics and Biotherapy, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, Xi'an, China
| | - Wuding Zhou
- Medical Research Council (MRC) Centre for Transplantation, King's College London, Guy's Hospital, London, United Kingdom
| | - Ke Li
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China.,National Local Joint Engineering Research Centre of Biodiagnostics and Biotherapy, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
33
|
Foley JF. Highlight: Antitumor strategies. Sci Signal 2017. [DOI: 10.1126/scisignal.aaq1397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Two papers highlight potential ways to boost the antitumor immune response.
Collapse
|