1
|
Bernardelli C, Caretti A, Lesma E. Dysregulated lipid metabolism in lymphangioleiomyomatosis pathogenesis as a paradigm of chronic lung diseases. Front Med (Lausanne) 2023; 10:1124008. [PMID: 36744130 PMCID: PMC9894443 DOI: 10.3389/fmed.2023.1124008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
A chronic inflammatory condition characterizes various lung diseases. Interestingly, a great contribution to inflammation is made by altered lipids metabolism, that can be caused by the deregulation of the mammalian target of rapamycin complex-1 (mTORC1) activity. There is evidence that one of mTOR downstream effectors, the sterol regulatory element-binding protein (SREBP), regulates the transcription of enzymes involved in the de novo fatty acid synthesis. Given its central role in cell metabolism, mTOR is involved in several biological processes. Among those, mTOR is a driver of senescence, a process that might contribute to the establishment of chronic lung disease because the characteristic irreversible inhibition of cell proliferation, associated to the acquisition of a pro-inflammatory senescence-associated secretory phenotype (SASP) supports the loss of lung parenchyma. The deregulation of mTORC1 is a hallmark of lymphangioleiomyomatosis (LAM), a rare pulmonary disease predominantly affecting women which causes cystic remodeling of the lung and progressive loss of lung function. LAM cells have senescent features and secrete SASP components, such as growth factors and pro-inflammatory molecules, like cancer cells. Using LAM as a paradigm of chronic and metastatic lung disease, here we review the published data that point out the role of dysregulated lipid metabolism in LAM pathogenesis. We will discuss lipids' role in the development and progression of the disease, to hypothesize novel LAM biomarkers and to propose the pharmacological regulation of lipids metabolism as an innovative approach for the treatment of the disease.
Collapse
Affiliation(s)
- Clara Bernardelli
- Laboratory of Pharmacology, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Anna Caretti
- Laboratory of Biochemistry and Molecular Biology, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Lesma
- Laboratory of Pharmacology, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy,*Correspondence: Elena Lesma,
| |
Collapse
|
2
|
Thomas A, Sumughan S, Dellacecca ER, Shivde RS, Lancki N, Mukhatayev Z, Vaca CC, Han F, Barse L, Henning SW, Zamora-Pineda J, Akhtar S, Gupta N, Zahid JO, Zack SR, Ramesh P, Jaishankar D, Lo AS, Moss J, Picken MM, Darling TN, Scholtens DM, Dilling DF, Junghans RP, Le Poole IC. Benign tumors in TSC are amenable to treatment by GD3 CAR T cells in mice. JCI Insight 2021; 6:152014. [PMID: 34806651 PMCID: PMC8663788 DOI: 10.1172/jci.insight.152014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/14/2021] [Indexed: 11/23/2022] Open
Abstract
Mutations underlying disease in tuberous sclerosis complex (TSC) give rise to tumors with biallelic mutations in TSC1 or TSC2 and hyperactive mammalian target of rapamycin complex 1 (mTORC1). Benign tumors might exhibit de novo expression of immunogens, targetable by immunotherapy. As tumors may rely on ganglioside D3 (GD3) expression for mTORC1 activation and growth, we compared GD3 expression in tissues from patients with TSC and controls. GD3 was overexpressed in affected tissues from patients with TSC and also in aging Tsc2+/– mice. As GD3 overexpression was not accompanied by marked natural immune responses to the target molecule, we performed preclinical studies with GD3 chimeric antigen receptor (CAR) T cells. Polyfunctional CAR T cells were cytotoxic toward GD3-overexpressing targets. In mice challenged with Tsc2–/– tumor cells, CAR T cells substantially and durably reduced the tumor burden, correlating with increased T cell infiltration. We also treated aged Tsc2+/– heterozygous (>60 weeks) mice that carry spontaneous Tsc2–/– tumors with GD3 CAR or untransduced T cells and evaluated them at endpoint. Following CAR T cell treatment, the majority of mice were tumor free while all control animals carried tumors. The outcomes demonstrate a strong treatment effect and suggest that targeting GD3 can be successful in TSC.
Collapse
Affiliation(s)
- Ancy Thomas
- Department of Dermatology, Feinberg School of Medicine.,Robert H. Lurie Comprehensive Cancer Center
| | | | | | | | - Nicola Lancki
- Quantitative Data Sciences Core, Robert H. Lurie Comprehensive Cancer Center; and
| | | | | | - Fei Han
- Department of Dermatology, Feinberg School of Medicine.,Robert H. Lurie Comprehensive Cancer Center
| | - Levi Barse
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Jesus Zamora-Pineda
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University, Maywood, Illinois, USA
| | - Suhail Akhtar
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University, Maywood, Illinois, USA
| | - Nikhilesh Gupta
- Robert H. Lurie Comprehensive Cancer Center.,Illinois Mathematics and Science Academy, Aurora, Illinois, USA
| | - Jasmine O Zahid
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University, Maywood, Illinois, USA
| | - Stephanie R Zack
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University, Maywood, Illinois, USA
| | | | | | - Agnes Sy Lo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Joel Moss
- Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Maria M Picken
- Department of Pathology, Loyola University, Maywood, Illinois, USA
| | - Thomas N Darling
- Department of Dermatology, School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| | - Denise M Scholtens
- Quantitative Data Sciences Core, Robert H. Lurie Comprehensive Cancer Center; and.,Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Daniel F Dilling
- Department of Medicine, Stritch School of Medicine, Loyola University, Maywood, Illinois, USA
| | - Richard P Junghans
- Department of Hematology/Oncology, School of Medicine, Boston University, Boston, Massachusetts, USA
| | - I Caroline Le Poole
- Department of Dermatology, Feinberg School of Medicine.,Robert H. Lurie Comprehensive Cancer Center.,Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
3
|
Immunotherapy for Lymphangioleiomyomatosis and Tuberous Sclerosis: Progress and Future Directions. Chest 2019; 156:1062-1067. [PMID: 31437431 DOI: 10.1016/j.chest.2019.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/24/2019] [Accepted: 08/04/2019] [Indexed: 01/19/2023] Open
Abstract
Pulmonary lymphangioleiomyomatosis (LAM) is a rare genetic multisystem disease characterized by the nodular proliferation of smooth muscle-like LAM cells, progressive cystic changes of the lung, lymphatic abnormalities, and renal angiomyolipomas (AMLs). LAM can arise sporadically or in women with the autosomal dominant disorder, tuberous sclerosis complex (TSC), in which hamartomatous tumors of brain, heart, skin, kidney, and lung are found. LAM and TSC are caused by mutations in the TSC1 or TSC2 tumor suppressor genes leading to elevated mechanistic/mammalian target of rapamycin complex activity. Recent data indicate that T cells within LAM nodules and renal AMLs exhibit features of T-cell exhaustion, with coinhibitory receptor programmed cell death protein 1 (PD-1) expression on tumor-infiltrating T cells. Treatment of animal models of TSC and LAM with anti-PD-1 antibodies or with the combination of anti-PD-1 and anti-CTLA4 antibodies has led to remarkable results, suppressing TSC2-null tumor growth and inducing tumor rejection. Here we review our current knowledge about the potential for immunotherapy for the treatment of LAM and TSC and highlight critical unknowns and key next steps.
Collapse
|
4
|
Maisel K, Merrilees MJ, Atochina-Vasserman EN, Lian L, Obraztsova K, Rue R, Vasserman AN, Zuo N, Angel LF, Gow AJ, Kang I, Wight TN, Eruslanov E, Swartz MA, Krymskaya VP. Immune Checkpoint Ligand PD-L1 Is Upregulated in Pulmonary Lymphangioleiomyomatosis. Am J Respir Cell Mol Biol 2018; 59:723-732. [PMID: 30095976 PMCID: PMC6293078 DOI: 10.1165/rcmb.2018-0123oc] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/21/2018] [Indexed: 12/29/2022] Open
Abstract
Pulmonary lymphangioleiomyomatosis (LAM) is a slow-progressing metastatic disease that is driven by mutations in the tumor suppressor tuberous sclerosis complex 1/2 (TSC1/2). Rapamycin inhibits LAM cell proliferation and is the only approved treatment, but it cannot cause the regression of existing lesions and can only stabilize the disease. However, in other cancers, immunotherapies such as checkpoint blockade against PD-1 and its ligand PD-L1 have shown promise in causing tumor regression and even curing some patients. Thus, we asked whether PD-L1 has a role in LAM progression. In vitro, PD-L1 expression in murine Tsc2-null cells is unaffected by mTOR inhibition with torin but can be upregulated by IFN-γ. Using immunohistochemistry and single-cell flow cytometry, we found increased PD-L1 expression both in human lung tissue from patients with LAM and in Tsc2-null lesions in a murine model of LAM. In this model, PD-L1 is highly expressed in the lung by antigen-presenting and stromal cells, and activated T cells expressing PD-1 infiltrate the affected lung. In vivo treatment with anti-PD-1 antibody significantly prolongs mouse survival in the model of LAM. Together, these data demonstrate that PD-1/PD-L1-mediated immunosuppression may occur in LAM, and suggest new opportunities for therapeutic targeting that may provide benefits beyond those of rapamycin.
Collapse
Affiliation(s)
- Katharina Maisel
- Institute for Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Mervyn J. Merrilees
- Department of Anatomy and Medical Imaging, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | | | - Lurong Lian
- Penn Center for Pulmonary Biology, Pulmonary, Allergy and Critical Care Division, and
| | - Kseniya Obraztsova
- Penn Center for Pulmonary Biology, Pulmonary, Allergy and Critical Care Division, and
| | - Ryan Rue
- Penn Center for Pulmonary Biology, Pulmonary, Allergy and Critical Care Division, and
| | | | - Ning Zuo
- Department of Anatomy and Medical Imaging, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Luis F. Angel
- New York University Langone Medical Center, New York, New York
| | - Andrew J. Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey; and
| | - Inkyung Kang
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington
| | - Thomas N. Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, Washington
| | - Evgeniy Eruslanov
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Melody A. Swartz
- Institute for Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Vera P. Krymskaya
- Penn Center for Pulmonary Biology, Pulmonary, Allergy and Critical Care Division, and
| |
Collapse
|
5
|
Pietrobon A, Delaney SP, Stanford WL. Could Immunotherapy Sink Its Teeth into Lymphangioleiomyomatosis? Am J Respir Cell Mol Biol 2018; 59:663-665. [DOI: 10.1165/rcmb.2018-0251ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Adam Pietrobon
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawa, Ontario, Canada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawa, Ontario, Canada
- Ottawa Institute of Systems BiologyOttawa, Ontario, Canada
| | - Sean P. Delaney
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawa, Ontario, Canada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawa, Ontario, Canada
- Ottawa Institute of Systems BiologyOttawa, Ontario, Canada
| | - William L. Stanford
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawa, Ontario, Canada
- Ottawa Institute of Systems BiologyOttawa, Ontario, Canada
- Department of Cellular and Molecular Medicineand
- Department of Biochemistry, Microbiology and ImmunologyUniversity of OttawaOttawa, Ontario, Canada
| |
Collapse
|
6
|
Liu HJ, Henske EP. Vitamin D binding protein: a new biomarker of disease severity in lymphangioleiomyomatosis. Eur Respir J 2018; 52:52/5/1801886. [PMID: 30385604 DOI: 10.1183/13993003.01886-2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 10/21/2018] [Indexed: 11/05/2022]
Affiliation(s)
- Heng-Jia Liu
- Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Elizabeth P Henske
- Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Liu J, Zheng X, Pang X, Li L, Wang J, Yang C, Du G. Ganglioside GD3 synthase (GD3S), a novel cancer drug target. Acta Pharm Sin B 2018; 8:713-720. [PMID: 30245960 PMCID: PMC6147802 DOI: 10.1016/j.apsb.2018.07.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/24/2018] [Accepted: 04/28/2018] [Indexed: 01/05/2023] Open
Abstract
Gangliosides are a class of important glycosphingolipids containing sialic acid that are widely distributed on the outer surface of cells and are abundantly distributed in brain tissue. Disialoganglioside with three glycosyl groups (GD3) and disialoganglioside with two glycosyl groups (GD2) are markedly increased in pathological conditions such as cancers and neurodegenerative diseases. GD3 and GD2 were found to play important roles in cancers by mediating cell proliferation, migration, invasion, adhesion, angiogenesis and in preventing immunosuppression of tumors. GD3 synthase (GD3S) is the regulatory enzyme of GD3 and GD2 synthesis, and is important in tumorigenesis and the development of cancers. The study of GD3S as a drug target may be of great significance for the discovery of new drugs for cancer treatment. This review will describe the gangliosides and their roles in physiological and pathological conditions; the roles of GD3 and GD2 in cancers; the expression, functions and mechanisms of GD3S, and its potential as a drug target in cancers.
Collapse
Affiliation(s)
- Jinyi Liu
- Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiangjin Zheng
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiaocong Pang
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Li Li
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Jinhua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Cui Yang
- Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
8
|
Julian LM, Delaney SP, Wang Y, Goldberg AA, Doré C, Yockell-Lelièvre J, Tam RY, Giannikou K, McMurray F, Shoichet MS, Harper ME, Henske EP, Kwiatkowski DJ, Darling TN, Moss J, Kristof AS, Stanford WL. Human Pluripotent Stem Cell-Derived TSC2-Haploinsufficient Smooth Muscle Cells Recapitulate Features of Lymphangioleiomyomatosis. Cancer Res 2017; 77:5491-5502. [PMID: 28830860 DOI: 10.1158/0008-5472.can-17-0925] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/22/2017] [Accepted: 08/16/2017] [Indexed: 01/06/2023]
Abstract
Lymphangioleiomyomatosis (LAM) is a progressive destructive neoplasm of the lung associated with inactivating mutations in the TSC1 or TSC2 tumor suppressor genes. Cell or animal models that accurately reflect the pathology of LAM have been challenging to develop. Here, we generated a robust human cell model of LAM by reprogramming TSC2 mutation-bearing fibroblasts from a patient with both tuberous sclerosis complex (TSC) and LAM (TSC-LAM) into induced pluripotent stem cells (iPSC), followed by selection of cells that resemble those found in LAM tumors by unbiased in vivo differentiation. We established expandable cell lines under smooth muscle cell (SMC) growth conditions that retained a patient-specific genomic TSC2+/- mutation and recapitulated the molecular and functional characteristics of pulmonary LAM cells. These include multiple indicators of hyperactive mTORC1 signaling, presence of specific neural crest and SMC markers, expression of VEGF-D and female sex hormone receptors, reduced autophagy, and metabolic reprogramming. Intriguingly, the LAM-like features of these cells suggest that haploinsufficiency at the TSC2 locus contributes to LAM pathology, and demonstrated that iPSC reprogramming and SMC lineage differentiation of somatic patient cells with germline mutations was a viable approach to generate LAM-like cells. The patient-derived SMC lines we have developed thus represent a novel cellular model of LAM that can advance our understanding of disease pathogenesis and develop therapeutic strategies against LAM. Cancer Res; 77(20); 5491-502. ©2017 AACR.
Collapse
Affiliation(s)
- Lisa M Julian
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada.,University of Ottawa, Ottawa, Ontario, Canada
| | - Sean P Delaney
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada.,University of Ottawa, Ottawa, Ontario, Canada.,Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| | - Ying Wang
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
| | | | - Carole Doré
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada
| | | | - Roger Y Tam
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada.,University of Ottawa, Ottawa, Ontario, Canada.,University of Toronto, Donnelly Centre for Cellular & Biomolecular Research, Boston, Massachusetts
| | - Krinio Giannikou
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Fiona McMurray
- University of Ottawa, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Health Sciences, Bethesda, Maryland
| | - Molly S Shoichet
- University of Toronto, Donnelly Centre for Cellular & Biomolecular Research, Boston, Massachusetts
| | - Mary-Ellen Harper
- University of Ottawa, Ottawa, Ontario, Canada.,Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Health Sciences, Bethesda, Maryland
| | - Elizabeth P Henske
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - David J Kwiatkowski
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thomas N Darling
- Uniformed Services University of Health Sciences, Bethesda, Maryland
| | - Joel Moss
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Arnold S Kristof
- Research Institute of McGill University Health Centre, Montreal, Quebec, Canada
| | - William L Stanford
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada. .,University of Ottawa, Ottawa, Ontario, Canada.,Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| |
Collapse
|
9
|
Eby JM, Barse L, Henning SW, Rabelink MJWE, Klarquist J, Gilbert ER, Hammer AM, Fernandez MF, Yung N, Khan S, Miller HG, Kessler ER, Garrett-Mayer E, Dilling DF, Hoeben RC, Le Poole IC. Alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 1 can support immune responses toward tumors overexpressing ganglioside D3 in mice. Cancer Immunol Immunother 2017; 66:63-75. [PMID: 27787577 PMCID: PMC11028533 DOI: 10.1007/s00262-016-1920-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/18/2016] [Indexed: 10/20/2022]
Abstract
An immunotherapeutic strategy is discussed supporting anti-tumor activity toward malignancies overexpressing ganglioside D3. GD3 can be targeted by NKT cells when derived moieties are presented in the context of CD1d. NKT cells can support anti-tumor responses by secreting inflammatory cytokines and through cytotoxicity toward CD1d+GD3+ tumors. To overexpress GD3, we generated expression vector DNA and an adenoviral vector encoding the enzyme responsible for generating GD3 from its ubiquitous precursor GM3. We show that DNA encoding α-N-acetyl-neuraminide α-2,8-sialyltransferase 1 (SIAT8) introduced by gene gun vaccination in vivo leads to overexpression of GD3 and delays tumor growth. Delayed tumor growth is dependent on CD1d expression by host immune cells, as shown in experiments engaging CD1d knockout mice. A trend toward greater NKT cell populations among tumor-infiltrating lymphocytes is associated with SIAT8 vaccination. A single adenoviral vaccination introduces anti-tumor activity similarly to repeated vaccination with naked DNA. Here, greater NKT tumor infiltrates were accompanied by marked overexpression of IL-17 in the tumor, later switching to IL-4. Our results suggest that a single intramuscular adenoviral vaccination introduces overexpression of GD3 by antigen-presenting cells at the injection site, recruiting NKT cells that provide an inflammatory anti-tumor environment. We propose adenoviral SIAT8 (AdV-SIAT8) can slow the growth of GD3 expressing tumors in patients.
Collapse
Affiliation(s)
- Jonathan M Eby
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
| | - Levi Barse
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Steven W Henning
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
| | - Martijn J W E Rabelink
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jared Klarquist
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
- Department of Immunology and Microbiology, University of Colorado Denver, Denver, CO, USA
| | - Emily R Gilbert
- Department of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Adam M Hammer
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
- Burn and Shock Trauma Institute, Loyola University Chicago, Maywood, IL, USA
| | - Manuel F Fernandez
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
| | - Nathan Yung
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA
| | - Safia Khan
- Oakton Community College, Des Plaines, IL, USA
| | | | - Edward R Kessler
- Department of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Elizabeth Garrett-Mayer
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Daniel F Dilling
- Department of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Rob C Hoeben
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - I Caroline Le Poole
- Oncology Research Institute, Loyola University Medical Center, Loyola University Chicago, Rm 203, 2160 S. 1st Avenue, Maywood, IL, 60153, USA.
- Departments of Pathology, Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA.
| |
Collapse
|
10
|
Lv J, Lv CQ, Mei P, Qi SM. Diagnosis value of membrane glycolipids biochemistry index in intracranial and gastrointestinal tumors. Asian Pac J Cancer Prev 2016; 16:2693-6. [PMID: 25854348 DOI: 10.7314/apjcp.2015.16.7.2693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The diagnostic value of membrane glycolipid biochemistry index, the lipid-bound sialic acid (LSA) and total sialic acid (TSA) in cerebrospinal fluid (CSF) was evaluated in 30 intracranial and 65 gastrointestinal tumors. The plasma LSA, TSA and red cell membrane sialic acid (R-SA) in were determined according to the method of Sevenmerhulm. Our results showed that the levels of LSA and TSA in CSF of intracranial tumor patients was higher than that of normal group(p<0.01). The concentration of TSA and LSA in patients with malignant glioma was higher than that of benign meningioma patients(P<0.01). No significance was found between intracranial halmatoma patients and normal control group for levels of membrane glycolipids (p>0.05). Results also found that the plasma LSA, TSA and R-SA of gastric carcinoma were significantly higher than those of control group (p<0.05); while no significant difference was found in the plasma LSA, TSA and R-SA levels between chronic gastritis, gastrohelcoma and normal control group (p>0.05). Plasma LSA, TSA and R-SA levels of gastric carcinoma patient were significantly higher than those of chronic gastritis patients and gastrohelcoma patients(p<0.05). It was also found that plasma LSA, TSA and R-SA contents were significantly higher in large intestine carcinoma patients than in benign in stestine tumor patients (p<0.05) while no significant difference was found between intestine benign tumor and normal control group (p>0.05). The levels of LSA, TSA and R-SA were obviously higher in the patients with metastasis than in the ones without (p<0.05.) The membrane glycolipid biochemistry index LSA and TSA in CSF are sensive markers for diagnosing intracranial tumors. For gastrointestinal malignant tumors the plasma LSA TSA and red blood cell membrane SA may be considered as auxiliary indicators for diagnosis. They can be used for distinguishing benign from malignant tumors.
Collapse
Affiliation(s)
- Jun Lv
- Department of Biochemistry, Wannan Medical College, Wuhu, Anhui, China E-mail :
| | | | | | | |
Collapse
|
11
|
Daniotti JL, Lardone RD, Vilcaes AA. Dysregulated Expression of Glycolipids in Tumor Cells: From Negative Modulator of Anti-tumor Immunity to Promising Targets for Developing Therapeutic Agents. Front Oncol 2016; 5:300. [PMID: 26779443 PMCID: PMC4703717 DOI: 10.3389/fonc.2015.00300] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 12/14/2015] [Indexed: 12/14/2022] Open
Abstract
Glycolipids are complex molecules consisting of a ceramide lipid moiety linked to a glycan chain of variable length and structure. Among these are found the gangliosides, which are sialylated glycolipids ubiquitously distributed on the outer layer of vertebrate plasma membranes. Changes in the expression of certain species of gangliosides have been described to occur during cell proliferation, differentiation, and ontogenesis. However, the aberrant and elevated expression of gangliosides has been also observed in different types of cancer cells, thereby promoting tumor survival. Moreover, gangliosides are actively released from the membrane of tumor cells, having a strong impact on impairing anti-tumor immunity. Beyond the undesirable effects of gangliosides in cancer cells, a substantial number of cancer immunotherapies have been developed in recent years that have used gangliosides as the main target. This has resulted in successful immune cell- or antibody-responses against glycolipids, with promising results having been obtained in clinical trials. In this review, we provide a general overview on the metabolism of glycolipids, both in normal and tumor cells, as well as examining glycolipid-mediated immune modulation and the main successes achieved in immunotherapies using gangliosides as molecular targets.
Collapse
Affiliation(s)
- Jose Luis Daniotti
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC, UNC-CONICET), Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba , Córdoba , Argentina
| | - Ricardo D Lardone
- Dirks/Dougherty Laboratory for Cancer Research, Department of Translational Immunology, John Wayne Cancer Institute at Providence Saint John's Health Center , Santa Monica, CA , USA
| | - Aldo A Vilcaes
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC, UNC-CONICET), Departamento de Química Biológica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba , Córdoba , Argentina
| |
Collapse
|
12
|
Lv J, Lv CQ, Xu L, Yang H. Plasma Content Variation and Correlation of Plasmalogen and GIS, TC, and TPL in Gastric Carcinoma Patients: A Comparative Study. Med Sci Monit Basic Res 2015; 21:157-60. [PMID: 26186056 PMCID: PMC4517849 DOI: 10.12659/msmbr.893908] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Background We studied the variation in plasma content of plasmalogen and ganglioside, total cholesterol (TC), and total phospholipid (TPL) in gastric carcinoma patients. The plasma plasmalogen levels were determined according to the vinyl ester bond method. Material/Methods Plasma ganglioside level was determined according to the method of Sevennerholm. The total cholesterols and total phospholipid were determined by routine methods. Results The plasma plasmalogen level of gastric carcinoma patients was significantly higher than in the control (normal) group, and the difference was markedly significant (p<0.01). The plasma total sialic acid (TSA) and lipid-bound sialic acid (LSA) of gastric carcinoma patients were higher than those of the normal control group (p<0.05). The total cholesterol content was higher than those in the normal control group (p<0.02), but the total phospholipid content was lower than in the normal control group and the difference was markedly significant (p<0.05). In the gastric carcinoma patients group, the plasmalogen and ganglioside-TSA levels were positively correlated (r=0.01, P<0.01). Plasmalogen and total cholesterols were also positively correlated (r=0.82, P<0.01), and plasmalogen and total phospholipid were negatively correlated (r=−0.82, p<0.01). Conclusions In gastric carcinoma patients, the plasma plasmalogen content was significantly elevated and was positively correlated with elevated level of gangliosides and total cholesterols, but it was negatively correlated with level of total phospholipids.
Collapse
Affiliation(s)
- Jun Lv
- Department of Biochemistry, Wannan Medical College, Wuhu, Anhui, China (mainland)
| | - Can-Qun Lv
- Department of Biochemistry, Wannan Medical College, Wuhu, Anhui, China (mainland)
| | - Lei Xu
- Department of Biochemistry, Wannan Medical College, Wuhu, Anhui, China (mainland)
| | - Hong Yang
- Department of Biochemistry, Wannan Medical College, Wuhu, Anhui, China (mainland)
| |
Collapse
|
13
|
Delaney SP, Julian LM, Stanford WL. The neural crest lineage as a driver of disease heterogeneity in Tuberous Sclerosis Complex and Lymphangioleiomyomatosis. Front Cell Dev Biol 2014; 2:69. [PMID: 25505789 PMCID: PMC4243694 DOI: 10.3389/fcell.2014.00069] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 11/02/2014] [Indexed: 12/20/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a rare neoplastic disease, best characterized by the formation of proliferative nodules that express smooth muscle and melanocytic antigens within the lung parenchyma, leading to progressive destruction of lung tissue and function. The pathological basis of LAM is associated with Tuberous Sclerosis Complex (TSC), a multi-system disorder marked by low-grade tumors in the brain, kidneys, heart, eyes, lung and skin, arising from inherited or spontaneous germ-line mutations in either of the TSC1 or TSC2 genes. LAM can develop either in a patient with TSC (TSC-LAM) or spontaneously (S-LAM), and it is clear that the majority of LAM lesions of both forms are characterized by an inactivating mutation in either TSC1 or TSC2, as in TSC. Despite this genetic commonality, there is considerable heterogeneity in the tumor spectrum of TSC and LAM patients, the basis for which is currently unknown. There is extensive clinical evidence to suggest that the cell of origin for LAM, as well as many of the TSC-associated tumors, is a neural crest cell, a highly migratory cell type with extensive multi-lineage potential. Here we explore the hypothesis that the types of tumors that develop and the tissues that are affected in TSC and LAM are dictated by the developmental timing of TSC gene mutations, which determines the identities of the affected cell types and the size of downstream populations that acquire a mutation. We further discuss the evidence to support a neural crest origin for LAM and TSC tumors, and propose approaches for generating humanized models of TSC and LAM that will allow cell of origin theories to be experimentally tested. Identifying the cell of origin and developing appropriate humanized models is necessary to truly understand LAM and TSC pathology and to establish effective and long-lasting therapeutic approaches for these patients.
Collapse
Affiliation(s)
- Sean P Delaney
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute Ottawa, ON, Canada ; Faculty of Graduate and Postdoctoral Studies, University of Ottawa Ottawa, ON, Canada ; Department of Cellular and Molecular Medicine, University of Ottawa Ottawa, ON, Canada
| | - Lisa M Julian
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute Ottawa, ON, Canada ; Faculty of Graduate and Postdoctoral Studies, University of Ottawa Ottawa, ON, Canada
| | - William L Stanford
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute Ottawa, ON, Canada ; Faculty of Graduate and Postdoctoral Studies, University of Ottawa Ottawa, ON, Canada ; Department of Cellular and Molecular Medicine, University of Ottawa Ottawa, ON, Canada ; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa Ottawa, ON, Canada
| |
Collapse
|