1
|
Zou B, Li M, Zhang J, Gao Y, Huo X, Li J, Fan Y, Guo Y, Liu X. Application of a risk score model based on glycosylation-related genes in the prognosis and treatment of patients with low-grade glioma. Front Immunol 2024; 15:1467858. [PMID: 39445005 PMCID: PMC11496118 DOI: 10.3389/fimmu.2024.1467858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Low-grade gliomas (LGG) represent a heterogeneous and complex group of brain tumors. Despite significant progress in understanding and managing these tumors, there are still many challenges that need to be addressed. Glycosylation, a common post-translational modification of proteins, plays a significant role in tumor transformation. Numerous studies have demonstrated a close relationship between glycosylation modifications and tumor progression. However, the biological function of glycosylation-related genes in LGG remains largely unexplored. Their potential roles within the LGG microenvironment are also not well understood. Methods We collected RNA-seq data and scRNA-seq data from patients with LGG from TCGA and GEO databases. The glycosylation pathway activity scores of each cluster and each patient were calculated by irGSEA and GSVA algorithms, and the differential genes between the high and low glycosylation pathway activity score groups were identified. Prognostic risk profiles of glycosylation-related genes were constructed using univariate Cox and LASSO regression analyses and validated in the CGGA database. Results An 8 genes risk score signature including ASPM, CHI3L1, LILRA4, MSN, OCIAD2, PTGER4, SERPING1 and TNFRSF12A was constructed based on the analysis of glycosylation-related genes. Patients with LGG were divided into high risk and low risk groups according to the median risk score. Significant differences in immunological characteristics, TIDE scores, drug sensitivity, and immunotherapy response were observed between these groups. Additionally, survival analysis of clinical medication information in the TCGA cohort indicated that high risk and low risk groups have different sensitivities to drug therapy. The risk score characteristics can thus guide clinical medication decisions for LGG patients. Conclusion Our study established glycosylation-related gene risk score signatures, providing new perspectives and approaches for prognostic prediction and treatment of LGG.
Collapse
Affiliation(s)
- Binbin Zou
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Mingtai Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jiachen Zhang
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yingzhen Gao
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoya Huo
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinhu Li
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yimin Fan
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanlin Guo
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaodong Liu
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
2
|
Knight A, Rihova L, Kralova R, Penka M, Adam Z, Pour L, Piskacek M, Hajek R. Plasmacytoid Dendritic Cells in Patients with MGUS and Multiple Myeloma. J Clin Med 2021; 10:jcm10163717. [PMID: 34442012 PMCID: PMC8396926 DOI: 10.3390/jcm10163717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/14/2021] [Accepted: 08/19/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Plasmacytoid dendritic cells (pDCs) play prominent roles in mediating innate and adaptive immune responses. However, it is unclear how pDCs contribute to the immunosuppressive tumor microenvironment described in multiple myeloma (MM). Methods: Newly diagnosed myeloma patients (MM, n = 37) were analyzed to determine the pDC counts in comparison to peripheral blood (PB, n = 53) and bone marrow (BM, n = 10) samples of age-matched healthy donors (HD) using flow cytometry. Second, proliferation of myeloma tumor cells in the presence of freshly isolated pDCs was examined. Third, production of IFNα by pDCs co-cultured with MM cells was determined by intracellular staining. Results: We found a highly significant reduction of circulating pDCs (p < 0.0001) and in bone marrow (p < 0.0001) of MM patients compared to HD. We also observed a significant decrease of pDCs (p = 0.004) in BM in patients with monoclonal gammopathy of undetermined significance (MGUS, n = 12). Importantly, we determined that pDCs promote proliferation specifically of MM cells and not the stromal cells and that pDCs secrete IFNα upon co-culture with MM tumor cells. Conclusions: Our results show altered pDC frequencies in the BM microenvironment in MGUS and MM patients at diagnosis. We showed the tumor-promoting function of pDCs that may mediate immune deficiencies affecting long-term disease control and treatment outcome.
Collapse
Affiliation(s)
- Andrea Knight
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic;
- Correspondence:
| | - Lucie Rihova
- Department of Hematology, University Hospital Brno, 625 00 Brno, Czech Republic; (L.R.); (R.K.); (M.P.)
| | - Romana Kralova
- Department of Hematology, University Hospital Brno, 625 00 Brno, Czech Republic; (L.R.); (R.K.); (M.P.)
| | - Miroslav Penka
- Department of Hematology, University Hospital Brno, 625 00 Brno, Czech Republic; (L.R.); (R.K.); (M.P.)
| | - Zdenek Adam
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, 625 00 Brno, Czech Republic; (Z.A.); (L.P.)
| | - Ludek Pour
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, 625 00 Brno, Czech Republic; (Z.A.); (L.P.)
| | - Martin Piskacek
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic;
| | - Roman Hajek
- Department of Hemato-Oncology, University Hospital Ostrava, 708 00 Ostrava, Czech Republic;
- Faculty of Medicine, University of Ostrava, 701 03 Ostrava, Czech Republic
| |
Collapse
|
3
|
Manouchehri N, Hussain RZ, Cravens PD, Esaulova E, Artyomov MN, Edelson BT, Wu GF, Cross AH, Doelger R, Loof N, Eagar TN, Forsthuber TG, Calvier L, Herz J, Stüve O. CD11c +CD88 +CD317 + myeloid cells are critical mediators of persistent CNS autoimmunity. Proc Natl Acad Sci U S A 2021; 118:e2014492118. [PMID: 33785592 PMCID: PMC8040603 DOI: 10.1073/pnas.2014492118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Natalizumab, a humanized monoclonal antibody (mAb) against α4-integrin, reduces the number of dendritic cells (DC) in cerebral perivascular spaces in multiple sclerosis (MS). Selective deletion of α4-integrin in CD11c+ cells should curtail their migration to the central nervous system (CNS) and ameliorate experimental autoimmune encephalomyelitis (EAE). We generated CD11c.Cre+/-ITGA4fl/fl C57BL/6 mice to selectively delete α4-integrin in CD11c+ cells. Active immunization and adoptive transfer EAE models were employed and compared with WT controls. Multiparameter flow cytometry was utilized to immunophenotype leukocyte subsets. Single-cell RNA sequencing was used to profile individual cells. α4-Integrin expression by CD11c+ cells was significantly reduced in primary and secondary lymphoid organs in CD11c.Cre+/-ITGA4fl/fl mice. In active EAE, a delayed disease onset was observed in CD11c.Cre+/-ITGA4fl/fl mice, during which CD11c+CD88+ cells were sequestered in the blood. Upon clinical EAE onset, CD11c+CD88+ cells appeared in the CNS and expressed CD317+ In adoptive transfer experiments, CD11c.Cre+/-ITGA4fl/fl mice had ameliorated clinical disease phenotype associated with significantly diminished numbers of CNS CD11c+CD88+CD317+ cells. In human cerebrospinal fluid from subjects with neuroinflammation, microglia-like cells display coincident expression of ITGAX (CD11c), C5AR1 (CD88), and BST2 (CD317). In mice, we show that only activated, but not naïve microglia expressed CD11c, CD88, and CD317. Finally, anti-CD317 treatment prior to clinical EAE substantially enhanced recovery in mice.
Collapse
Affiliation(s)
- Navid Manouchehri
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Rehana Z Hussain
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Petra D Cravens
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Ekaterina Esaulova
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Maxim N Artyomov
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Brian T Edelson
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gregory F Wu
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Anne H Cross
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Richard Doelger
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Nicolas Loof
- The Moody Foundation Flow Cytometry Facility, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Todd N Eagar
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030
| | - Thomas G Forsthuber
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249
| | - Laurent Calvier
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs University, 79085 Freiburg, Germany
| | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390;
- Neurology Section, VA North Texas Health Care System, Dallas, TX 75216
| |
Collapse
|
4
|
Falco M, Palma G, Rea D, De Biase D, Scala S, D'Aiuto M, Facchini G, Perdonà S, Barbieri A, Arra C. Tumour biomarkers: homeostasis as a novel prognostic indicator. Open Biol 2016; 6:160254. [PMID: 27927793 PMCID: PMC5204124 DOI: 10.1098/rsob.160254] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/10/2016] [Indexed: 12/15/2022] Open
Abstract
The term 'personalized medicine' refers to a medical procedure that consists in the grouping of patients based on their predicted individual response to therapy or risk of disease. In oncologic patients, a 'tailored' therapeutic approach may potentially improve their survival and well-being by not only reducing the tumour, but also enhancing therapeutic response and minimizing the adverse effects. Diagnostic tests are often used to select appropriate and optimal therapies that rely both on patient genome and other molecular/cellular analysis. Several studies have shown that lifestyle and environmental factors can influence the epigenome and that epigenetic events may be involved in carcinogenesis. Thus, in addition to traditional biomarkers, epigenetic factors are raising considerable interest, because they could potentially be used as an excellent tool for cancer diagnosis and prognosis. In this review, we summarize the role of conventional cancer genetic biomarkers and their association with epigenomics. Furthermore, we will focus on the so-called 'homeostatic biomarkers' that result from the physiological response to cancer, emphasizing the concept that an altered 'new' homeostasis influence not only tumour environment, but also the whole organism.
Collapse
Affiliation(s)
- Michela Falco
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Giuseppe Palma
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Domenica Rea
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Davide De Biase
- Department of Veterinary Medicine and Animal Production, University of Naples 'Federico II', Via Delpino 1, 80137 Naples, Italy
| | - Stefania Scala
- Molecular lmmunology and Immuneregulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, IRCCS Naples 'Fondazione G. Pascale', Naples, italy, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Massimiliano D'Aiuto
- Division of Breast Surgery, Department of Breast Disease, National Cancer Institute, IRCCS, 'Fondazione Pascale', Naples, Italy
| | - Gaetano Facchini
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, , Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione G. Pascale', IRCCS, 80131 Naples, Italy
| | - Sisto Perdonà
- Department of Urology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione G. Pascale', IRCCS, 80131 Naples, Italy
| | - Antonio Barbieri
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Claudio Arra
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| |
Collapse
|
5
|
Extracting tumor tissue immune status from expression profiles: correlating renal cancer prognosis with tumor-associated immunome. Oncotarget 2016; 6:33191-205. [PMID: 26384298 PMCID: PMC4741758 DOI: 10.18632/oncotarget.5052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 08/28/2015] [Indexed: 01/25/2023] Open
Abstract
Investigating the expression of genes in cancer-associated immune cells (immunome) is imperative for prognosis prediction. However, evaluating the expression of immune-associated genes within cancer biopsy is subject to significant inconsistencies related to the sampling methodology. Here, we present immFocus, a method for extracting immune signals from total RNA sequencing of tumor biopsies, intended for immunity depiction and prognosis evaluation. It is based on reducing the variation which biopsy preparation adds to the apparent expression levels of immune genes. We employed immFocus to normalize gene expression with an immune index using data obtained from renal clear cell carcinoma biopsies. Genes that became less variable due to normalization were found to be preferentially immune-related. Moreover, immune-related genes tended to become more prognostic due to the normalization. These results demonstrate, for the first time, that whole transcriptome sequencing can be used for interrogation of a cancer immunome and for advancing immune-based prognosis.
Collapse
|
6
|
Jordan M, Waxman DJ. CpG-1826 immunotherapy potentiates chemotherapeutic and anti-tumor immune responses to metronomic cyclophosphamide in a preclinical glioma model. Cancer Lett 2015; 373:88-96. [PMID: 26655275 DOI: 10.1016/j.canlet.2015.11.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/26/2015] [Accepted: 11/27/2015] [Indexed: 02/08/2023]
Abstract
Cyclophosphamide administered on an intermittent metronomic schedule induces strong immune-dependent regression in several glioma models. Here we investigate whether this immunogenic chemotherapy can be potentiated by combination with the immune stimulatory TLR9 agonist CpG-1826. CpG-1826 treatment of GL261 gliomas implanted in immune competent mice induced tumor growth delay associated with increased tumor recruitment of macrophages and B cells. Anti-tumor responses varied between individuals, with CpG-1826 inducing robust tumor growth delay in ~50% of treated mice. Both high and low CpG-1826-responsive mice showed striking improvements when CpG-1826 was combined with cyclophosphamide treatment. Tumor-associated macrophages, B cells, dendritic cells, and cytotoxic T cells were increased, T regulatory cells were not induced, and long-term GL261 glioma regression with immune memory was achieved when CpG-1826 was combined with either single cyclophosphamide dosing (90 mg/kg) or metronomic cyclophosphamide treatment (two cycles at 45 mg/kg, spaced 12-days apart). B16F10 melanoma, a low immunogenic tumor model, also showed enhanced immune and anti-tumor responses to cyclophosphamide/CpG-1826 chemoimmunotherapy, but unlike GL261 tumors, did not regress. TLR9-based immunotherapy can thus be effectively combined with immunogenic cyclophosphamide treatment to enhance immune-based anti-tumor responses, even in poorly immunogenic cancer models.
Collapse
Affiliation(s)
- Marie Jordan
- Division of Cell and Molecular Biology, Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - David J Waxman
- Division of Cell and Molecular Biology, Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA.
| |
Collapse
|
7
|
Plasmacytoid dendritic cells in the tumor microenvironment: immune targets for glioma therapeutics. Neoplasia 2013; 14:757-70. [PMID: 22952428 DOI: 10.1593/neo.12794] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 07/10/2012] [Accepted: 07/11/2012] [Indexed: 01/19/2023] Open
Abstract
Adenovirus-mediated delivery of the immune-stimulatory cytokine Flt3L and the conditionally cytotoxic thymidine kinase (TK) induces tumor regression and long-term survival in preclinical glioma (glioblastoma multiforme [GBM]) models. Flt3L induces expansion and recruitment of plasmacytoid dendritic cells (pDCs) into the brain. Although pDCs can present antigen and produce powerful inflammatory cytokines, that is, interferon α (IFN-α), their role in tumor immunology remains debated. Thus, we studied the role of pDCs and IFN-α in Ad.TK/GCV+ Ad.Flt3L-mediated anti-GBM therapeutic efficacy. Our data indicate that the combined gene therapy induced recruitment of plasmacytoid DCs (pDCs) into the tumor mass; which were capable of in vivo phagocytosis, IFN-α release, and T-cell priming. Thus, we next used either pDCs or an Ad vector encoding IFN-α delivered within the tumor microenvironment. When rats were treated with Ad.TK/GCV in combination with pDCs or Ad-IFN-α, they exhibited 35% and 50% survival, respectively. However, whereas intracranial administration of Ad.TK/GCV + Ad.Flt3L exhibited a high safety profile, Ad-IFN-α led to severe local inflammation, with neurologic and systemic adverse effects. To elucidate whether the efficacy of the immunotherapy was dependent on IFN-α-secreting pDCs, we administered an Ad vector encoding B18R, an IFN-α antagonist, which abrogated the antitumoral effect of Ad.TK/GCV + Ad.Flt3L. Our data suggest that IFN-α release by activated pDCs plays a critical role in the antitumor effect mediated by Ad.TK/GCV + Ad.Flt3L. In summary, taken together, our results demonstrate that pDCs mediate anti-GBM therapeutic efficacy through the production of IFN-α, thus manipulation of pDCs constitutes an attractive new therapeutic target for the treatment of GBM.
Collapse
|