1
|
Skurikhin E, Zhukova M, Ermakova N, Pan E, Widera D, Sandrikina L, Kogai L, Kushlinskii N, Kubatiev A, Morozov S, Dygai A. Age-related features of lung cancer treatment using reprogrammed CD8 positive T cells in mice subjected to injection of Lewis lung carcinoma cells. Thorac Cancer 2024; 15:2000-2020. [PMID: 39169897 PMCID: PMC11444928 DOI: 10.1111/1759-7714.15426] [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: 06/24/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Awareness of age-related features of carcinogenesis and the importance of cellular immunity is crucial for developing effective antitumor therapies for specific patient groups. METHODS In this study, we examined different populations of cancer stem cells (CSCs) and circulating tumor cells (CTCs) in "young" (8-10 weeks) and "aged" (80-82 weeks) C57BL/6 male mice. We used an orthotopic model of Lewis lung carcinoma (LLC) to evaluate the effectiveness of cell therapy targeting lung cancer through reprogrammed CD8-positive T cells (rCD8+ T cells) in mice from two different ages. RESULTS The findings revealed that tumor progression with age is primarily caused by impaired recruitment of T cells to the lungs. Additionally, a lower number of CTCs and CSCs were observed in younger mice compared to the older mice. The antitumor effect of rCD8+ T cells in aged mice was found to be inferior to that in young mice, which can be attributed to the reduced impact of therapy on specific CSCs populations. CONCLUSIONS These results offer new insights into the treatment of lung cancer using rCD8+ T cells. Considering the age-related characteristics influencing disease progression, this therapy has the potential to significantly enhance the effectiveness of treatment methods.
Collapse
Affiliation(s)
| | - Mariia Zhukova
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Natalia Ermakova
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Edgar Pan
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, Whiteknights Campus, Reading, UK
| | - Lubov Sandrikina
- Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Lena Kogai
- Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
- Ministry of Health of the Russian Federation, Siberian State Medical University, Tomsk, Russia
| | | | - Aslan Kubatiev
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Sergey Morozov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Alexander Dygai
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
2
|
Pershina OV, Ermakova NN, Pakhomova AV, Pan ES, Sandrikina LA, Zhukova MA, Kogai LV, Dygai AM, Skurikhin EG. Age-Related Features of the Response of Cancer Stem Cells and T Cells in Experimental Lung Cancer. Bull Exp Biol Med 2024; 176:486-490. [PMID: 38492106 DOI: 10.1007/s10517-024-06052-9] [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: 07/10/2023] [Indexed: 03/18/2024]
Abstract
The responses of tumor stem cells and various populations of CD4 and CD8 T cells of young and aged C57BL/6 mice were studied in a lung cancer model. Using Lewis lung carcinoma cell line, an orthotopic model of lung cancer was modeled. Cancer stem cells, circulating tumor cells, and various populations of CD4 and CD8 T cells in the blood and lung tissue were studied by cytometry. We revealed age-related differences in the content of various populations of CD4 and CD8 T cells in the blood and lungs of intact young and aged mice. Age-related features of the reaction of various populations of cancer stem cells and CD4 and CD8 T cells in the blood and lungs of animals in the Lewis lung carcinoma were shown.
Collapse
Affiliation(s)
- O V Pershina
- Laboratory of Regenerative Pharmacology, E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia.
| | - N N Ermakova
- Laboratory of Regenerative Pharmacology, E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - A V Pakhomova
- Laboratory of Regenerative Pharmacology, E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - E S Pan
- Laboratory of Regenerative Pharmacology, E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - L A Sandrikina
- Laboratory of Regenerative Pharmacology, E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - M A Zhukova
- Laboratory of Regenerative Pharmacology, E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - L V Kogai
- Laboratory of Regenerative Pharmacology, E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - A M Dygai
- Laboratory of Regenerative Pharmacology, E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
- Research Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - E G Skurikhin
- Research Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
3
|
Singh A, Sharma A. Lymphoid tissue inducer cells in cancer: a potential therapeutic target. Mol Cell Biochem 2023; 478:2789-2794. [PMID: 36922480 DOI: 10.1007/s11010-023-04699-y] [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: 10/25/2022] [Accepted: 03/02/2023] [Indexed: 03/17/2023]
Abstract
Tumor cells are dynamic in nature; these cells first acquire immune surveillance and then escape from the immune system. Hence, progressed cancer cells distribute and metastasize to other organs via blood vessels as well as from the lymphatic system. Prognosis and treatment of metastatic cancer patients remain a major challenge nowadays. Till now, lots of target -based and immune checkpoint blocker therapies are used to treat disease patients. But these therapies fail to control the dissemination and metastasis of cancer. Before designing a treatment regimen for metastatic patients, understanding the mechanism of tumor cells spreading within lymph vessels remain undetermined. Construction of lymphoid structures since embryonic to adult stage are depend upon LTi. Foundation of lymph node, payer patches and TLO is initiated and regulated through these cells in any part of the body. During tumor growth, newly developed lymph node contained MDSCs and Treg cells which inhibit the immune response and promote tumor invasion and metastasis. LTi reconstituted lymph node can be used for both early and high risk detection of cancers. High and low risk of tumor growth and invasion depend upon the location and composition of immune cells within lymph nodes. However, LTi are not reported as predictive marker in cancer till date. Recent reports in cancer indicate that LTi cells are engaged in the spreading of tumor cells into a lymphatic vessel. Through this review we are trying to brief the development and role of the LTi in immune system during homeostasis and cancer.
Collapse
Affiliation(s)
- Ashu Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
4
|
Gray S, Ottensmeier CH. Advancing Understanding of Non-Small Cell Lung Cancer with Multiplexed Antibody-Based Spatial Imaging Technologies. Cancers (Basel) 2023; 15:4797. [PMID: 37835491 PMCID: PMC10571797 DOI: 10.3390/cancers15194797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a cause of significant morbidity and mortality, despite significant advances made in its treatment using immune checkpoint inhibitors (ICIs) over the last decade; while a minority experience prolonged responses with ICIs, benefit is limited for most patients. The development of multiplexed antibody-based (MAB) spatial tissue imaging technologies has revolutionised analysis of the tumour microenvironment (TME), enabling identification of a wide range of cell types and subtypes, and analysis of the spatial relationships and interactions between them. Such study has the potential to translate into a greater understanding of treatment susceptibility and resistance, factors influencing prognosis and recurrence risk, and identification of novel therapeutic approaches and rational treatment combinations to improve patient outcomes in the clinic. Herein we review studies that have leveraged MAB technologies to deliver novel insights into the TME of NSCLC.
Collapse
Affiliation(s)
- Simon Gray
- Department of Molecular and Clinical Cancer Medicine, Faculty of Health and Life Sciences, University of Liverpool, Ashton St., Liverpool L69 3GB, UK
- Department of Medical Oncology, The Clatterbridge Cancer Centre NHS Foundation Trust, Pembroke Pl., Liverpool L7 8YA, UK
| | - Christian H. Ottensmeier
- Department of Molecular and Clinical Cancer Medicine, Faculty of Health and Life Sciences, University of Liverpool, Ashton St., Liverpool L69 3GB, UK
- Department of Medical Oncology, The Clatterbridge Cancer Centre NHS Foundation Trust, Pembroke Pl., Liverpool L7 8YA, UK
| |
Collapse
|
5
|
Li R, Lin Y, Wang Y, Wang S, Yang Y, Mu X, Chen Y, Gao Z. Characterization of the Tumor Immune Microenvironment in Lung Squamous Cell Carcinoma Using Imaging Mass Cytometry. Front Oncol 2021; 11:620989. [PMID: 33869005 PMCID: PMC8047498 DOI: 10.3389/fonc.2021.620989] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 03/15/2021] [Indexed: 12/25/2022] Open
Abstract
Background Lung squamous cell carcinoma (LUSC) is a major subtype of non-small cell lung cancer. The tumor immune microenvironment (TIME) affects the anti-tumor immune response and the patient’s prognosis, although the TIME in LUSC patients is incompletely understood. Methods We retrospectively collected surgical specimens from patients with previously untreated primary LUSC. Histopathological examination was used to identify tumor regions and adjacent regions, and imaging mass cytometry was used to characterize the immune cells in those regions. The results were compared between regions and between patients. Results We identified heterogeneity in the TIME on comparing different patients with LUSC, although the tumor region and adjacent region both exhibited an immune response to the tumor. The TIME typically included a large number of infiltrating and activated T-cells, especially CD8+ T-cells, which closely interacted with the tumor cells in the tumor region. There was limited infiltration of B-cells, NK cells, and NKT cells, while the major immune suppressor cells were CD33+ myeloid-derived cells. We also identified a novel population of CD3−CD4+ cells with high expression of Foxp3 and TNFα, which might modulate the tumor microenvironment and play a proinflammatory role in the TIME. Conclusions The TIME of LUSC appears to be immunogenic and heterogenous, with predominant infiltration of activated CD8+ T-cells. The interactions between the tumor cells and T-cells facilitate the anti-tumor activity. A novel subpopulation of CD3−CD4+ cells with high TNFα and Foxp3 expression may modulate the tumor microenvironment and play a proinflammatory role.
Collapse
Affiliation(s)
- Ran Li
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Ying Lin
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Yu Wang
- Life Science Institute, Jinzhou Medical University, Jinzhou, China
| | - Shaoyuan Wang
- Beijing Gencode Diagnostics Laboratory, Beijing, China
| | - Yang Yang
- Beijing Gencode Diagnostics Laboratory, Beijing, China
| | - Xinlin Mu
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Yusheng Chen
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, China.,Department of Respiratory and Critical Care Medicine, Fujian Provincial Hospital, Fuzhou, China
| | - Zhancheng Gao
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| |
Collapse
|
6
|
Meininger I, Carrasco A, Rao A, Soini T, Kokkinou E, Mjösberg J. Tissue-Specific Features of Innate Lymphoid Cells. Trends Immunol 2020; 41:902-917. [PMID: 32917510 DOI: 10.1016/j.it.2020.08.009] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023]
Abstract
Although the function of the circulating immune cell compartment has been studied in detail for decades, limitations in terms of access and cell yields from peripheral tissues have restricted our understanding of tissue-based immunity, particularly in humans. Recent advances in high-throughput protein analyses, transcriptional profiling, and epigenetics have partially overcome these obstacles. Innate lymphoid cells (ILCs) are predominantly tissue-resident, and accumulating data indicate that they have significant tissue-specific functions. We summarize current knowledge of ILC phenotypes in various tissues in mice and humans, aiming to clarify ILC immunity in distinct anatomical locations.
Collapse
Affiliation(s)
- Isabel Meininger
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Carrasco
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Rao
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tea Soini
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Efthymia Kokkinou
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jenny Mjösberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
7
|
Gupta A, Kumar D, Puri S, Puri V. Neuroimmune Mechanisms in Signaling of Pain During Acute Kidney Injury (AKI). Front Med (Lausanne) 2020; 7:424. [PMID: 32850914 PMCID: PMC7427621 DOI: 10.3389/fmed.2020.00424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 07/01/2020] [Indexed: 11/18/2022] Open
Abstract
Acute kidney injury (AKI) is a significant global health concern. The primary causes of AKI include ischemia, sepsis and nephrotoxicity. The unraveled interface between nervous system and immune response with specific focus on pain pathways is generating a huge interest in reference to AKI. The nervous system though static executes functions by nerve fibers throughout the body. Neuronal peptides released by nerves effect the immune response to mediate the hemodynamic system critical to the functioning of kidney. Pain is the outcome of cellular cross talk between nervous and immune systems. The widespread release of neuropeptides, neurotransmitters and immune cells contribute to bidirectional neuroimmune cross talks for pain manifestation. Recently, we have reported pain pathway genes that may pave the way to better understand such processes during AKI. An auxiliary understanding of the functions and communications in these systems will lead to novel approaches in pain management and treatment through the pathological state, specifically during acute kidney injury.
Collapse
Affiliation(s)
- Aprajita Gupta
- Centre for Systems Biology & Bioinformatics, Panjab University, Chandigarh, India
| | - Dev Kumar
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sanjeev Puri
- Department of Biotechnology, University Institute of Engineering and Technology, Panjab University, Chandigarh, India
| | - Veena Puri
- Centre for Systems Biology & Bioinformatics, Panjab University, Chandigarh, India
| |
Collapse
|
8
|
Hu Z, Wang W, Thakral B, Chen Z, Estrov Z, Bueso-Ramos CE, Verstovsek S, Medeiros LJ, Wang SA. Lymphocytic variant of hypereosinophilic syndrome: A report of seven cases from a single institution. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 100:352-360. [PMID: 32157815 DOI: 10.1002/cyto.b.21874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/09/2020] [Accepted: 02/26/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Lymphocytic variant of hypereosinophilic syndrome (L-HES) is a subtype of HES driven by cytokines produced by clonal T-cells. Due to the rarity of its occurrence and challenges in diagnosis, this subtype of HES is under recognized. METHODS AND RESULTS We report seven patients with L-HES, diagnosed from a group of 136 patients who were referred to our institution for the work-up of hypereosinophilia. The clinical presentation, symptoms and signs were heterogeneous and uncharacteristic; indistinguishable from idiopathic HES. Flow cytometry immunophenotypic analysis revealed aberrant T-cells in all patients, with a Th2 immunophenotype, CD2 + CD3-CD4 + CD5 + CD7dim+/-CD8- in six of seven (86%) cases. CD10 was partially expressed in one of seven (14%) cases, and clonal TCR gene rearrangement was detected by PCR in five of seven (71%) patients. All patients were treated with corticosteroids and two of seven (29%) patients received anti-IL5 antibody therapy. With a median follow-up time of 7.5 years (2.3-14.1 years), one (11%) patient developed peripheral T-cell lymphoma 6.1 years after the initial diagnosis of L-HES and responded well to chemotherapy. All patients were alive at the last follow-up. CONCLUSION In conclusion, a combination of flow cytometry immunophenotyping and molecular analysis allows the identification of aberrant T-cells, facilitating a diagnosis of L-HES in patients with eosinophilia. A correct diagnosis is essential for the proper management of these patients.
Collapse
Affiliation(s)
- Zhihong Hu
- Department of Pathology, The University of Texas Health Center at Houston, Houston, Texas, USA
| | - Wei Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Beenu Thakral
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhining Chen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zeev Estrov
- Department of Pathology, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
| | - Carlos E Bueso-Ramos
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Srdan Verstovsek
- Department of Pathology, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sa A Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
9
|
Anoveros-Barrera A, Bhullar AS, Stretch C, Dunichand-Hoedl AR, Martins KJB, Rieger A, Bigam D, McMullen T, Bathe OF, Putman CT, Field CJ, Baracos VE, Mazurak VC. Immunohistochemical phenotyping of T cells, granulocytes, and phagocytes in the muscle of cancer patients: association with radiologically defined muscle mass and gene expression. Skelet Muscle 2019; 9:24. [PMID: 31521204 PMCID: PMC6744687 DOI: 10.1186/s13395-019-0209-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/16/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inflammation is a recognized contributor to muscle wasting. Research in injury and myopathy suggests that interactions between the skeletal muscle and immune cells confer a pro-inflammatory environment that influences muscle loss through several mechanisms; however, this has not been explored in the cancer setting. This study investigated the local immune environment of the muscle by identifying the phenotype of immune cell populations in the muscle and their relationship to muscle mass in cancer patients. METHODS Intraoperative muscle biopsies were collected from cancer patients (n = 30, 91% gastrointestinal malignancies). Muscle mass was assessed histologically (muscle fiber cross-sectional area, CSA; μm2) and radiologically (lumbar skeletal muscle index, SMI; cm2/m2 by computed tomography, CT). T cells (CD4 and CD8) and granulocytes/phagocytes (CD11b, CD14, and CD15) were assessed by immunohistochemistry. Microarray analysis was conducted in the muscle of a second cancer patient cohort. RESULTS T cells (CD3+), granulocytes/phagocytes (CD11b+), and CD3-CD4+ cells were identified. Muscle fiber CSA (μm2) was positively correlated (Spearman's r = > 0.45; p = < 0.05) with the total number of T cells, CD4, and CD8 T cells and granulocytes/phagocytes. In addition, patients with the smallest SMI exhibited fewer CD8 T cells within their muscle. Consistent with this, further exploration with gene correlation analyses suggests that the presence of CD8 T cells is negatively associated (Pearson's r = ≥ 0.5; p = <0.0001) with key genes within muscle catabolic pathways for signaling (ACVR2B), ubiquitin proteasome (FOXO4, TRIM63, FBXO32, MUL1, UBC, UBB, UBE2L3), and apoptosis/autophagy (CASP8, BECN1, ATG13, SIVA1). CONCLUSION The skeletal muscle immune environment of cancer patients is comprised of immune cell populations from the adaptive and innate immunity. Correlations of T cells, granulocyte/phagocytes, and CD3-CD4+ cells with muscle mass measurements indicate a positive relationship between immune cell numbers and muscle mass status in cancer patients. Further exploration with gene correlation analyses suggests that the presence of CD8 T cells is negatively correlated with components of muscle catabolism.
Collapse
Affiliation(s)
- Ana Anoveros-Barrera
- Department of Agricultural, Food & Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, 4-002 Li Ka Shing Centre, Edmonton, Alberta, T6G 2P5, Canada
| | - Amritpal S Bhullar
- Department of Agricultural, Food & Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, 4-002 Li Ka Shing Centre, Edmonton, Alberta, T6G 2P5, Canada
| | - Cynthia Stretch
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Abha R Dunichand-Hoedl
- Department of Agricultural, Food & Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, 4-002 Li Ka Shing Centre, Edmonton, Alberta, T6G 2P5, Canada
| | - Karen J B Martins
- Department of Agricultural, Food & Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, 4-002 Li Ka Shing Centre, Edmonton, Alberta, T6G 2P5, Canada
| | - Aja Rieger
- Flow Cytometry Facility, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - David Bigam
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Todd McMullen
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Oliver F Bathe
- Department of Oncology and Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - Charles T Putman
- Faculty of Kinesiology, Sport, and Recreation, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Catherine J Field
- Department of Agricultural, Food & Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, 4-002 Li Ka Shing Centre, Edmonton, Alberta, T6G 2P5, Canada
| | - Vickie E Baracos
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Vera C Mazurak
- Department of Agricultural, Food & Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, 4-002 Li Ka Shing Centre, Edmonton, Alberta, T6G 2P5, Canada.
| |
Collapse
|
10
|
Chan TY, Yen CL, Huang YF, Lo PC, Nigrovic PA, Cheng CY, Wang WZ, Wu SY, Shieh CC. Increased ILC3s associated with higher levels of IL-1β aggravates inflammatory arthritis in mice lacking phagocytic NADPH oxidase. Eur J Immunol 2019; 49:2063-2073. [PMID: 31350760 DOI: 10.1002/eji.201948141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/27/2019] [Accepted: 07/23/2019] [Indexed: 01/12/2023]
Abstract
The role of redox regulation in immune-mediated arthritis has been previously described. However, the relationship between innate immune cells, including innate lymphoid cells (ILCs) and phagocyte-derived ROS, in this process remains unclear. Here, we characterize ILCs and measure the IL-1 family cytokines along with other cytokines relevant to ILC functions and development in serum-induced arthritic joints in wild type and phagocytic NADPH oxidase (NOX2)-deficient Ncf1-/- mice. We found more severe serum-induced joint inflammation and increased NCR+ ILC3s in inflamed joints of Ncf1-/- mice. Furthermore, in vitro stimulation with IL-1β on Tbet+ ILC1s from joints facilitated their differentiation into ROR-γt+ ILC3s. Moreover, treatment with IL-1 antagonists effectively lowered the proportions of NCR+ ILC3s and IL-17A producing ILC3s in Ncf1-/- arthritic mice and ameliorated the joint inflammation. These results suggest that NOX2 is an essential regulator of ILC transdifferentiation and may mediate this process in a redox-dependent manner through IL-1β production in the inflammatory joint. Our findings shed important light on the role of ILCs in the initiation and progression in tissue inflammation and delineate a novel innate immune cell-mediated pathogenic mechanism through which redox regulation may determine the direction of immune responses in joints.
Collapse
Affiliation(s)
- Tzu-Yi Chan
- Institute of Clinical Medicine, National Cheng-Kung University Medical College, Tainan, Taiwan
| | - Chia-Liang Yen
- Institute of Clinical Medicine, National Cheng-Kung University Medical College, Tainan, Taiwan
| | - Ya-Fang Huang
- National Laboratory Animal Center, National Applied Research Laboratories, Tainan, Taiwan
| | - Pei-Chi Lo
- Division of Organ Transplantation, Department of Surgery, Osaka University. Graduate School of Medicine, Osaka, Japan
| | - Peter A Nigrovic
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA, USA.,Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Chia-Ying Cheng
- Institute of Clinical Medicine, National Cheng-Kung University Medical College, Tainan, Taiwan
| | - Wei-Zhi Wang
- Institute of Clinical Medicine, National Cheng-Kung University Medical College, Tainan, Taiwan
| | - Szu-Yu Wu
- Institute of Clinical Medicine, National Cheng-Kung University Medical College, Tainan, Taiwan
| | - Chi-Chang Shieh
- Institute of Clinical Medicine, National Cheng-Kung University Medical College, Tainan, Taiwan.,Department of Pediatrics, National Cheng-Kung University Hospital, Tainan, Taiwan
| |
Collapse
|
11
|
Yoshitomi H. Regulation of Immune Responses and Chronic Inflammation by Fibroblast-Like Synoviocytes. Front Immunol 2019; 10:1395. [PMID: 31275325 PMCID: PMC6593115 DOI: 10.3389/fimmu.2019.01395] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 06/03/2019] [Indexed: 11/13/2022] Open
Abstract
Synovial tissue is a membranous non-immune organ lining joint cavities where it supports local immune responses, and functions directly and indirectly in joint destruction due to chronic inflammatory diseases such as rheumatoid arthritis (RA). Fibroblast-like synoviocytes (FLS), the dominant non-immune cells of synovial tissues, mainly contribute to joint destruction via multiple mechanisms. In RA, FLS respond to endogenous ligands of pattern recognition receptors (PRRs) and inflammatory cytokines as non-immune cells. In addition, FLS aid in the activation of immune responses by interacting with immune cells and by supporting ectopic lymphoid-like structure (ELS) formation in synovial tissues. Moreover, FLS directly cause the pathogenicity of RA i.e., joint deformities. Here, we describe new findings and review the mechanisms underlying the regulation of immune reactions by non-immune FLS and their roles in inflammatory diseases such as RA.
Collapse
Affiliation(s)
- Hiroyuki Yoshitomi
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
12
|
Affiliation(s)
- Yannick Simoni
- Agency for Science, Technology and Research (A(∗)STAR), Singapore Immunology Network (SIgN), 138648 Singapore.
| | - Evan W Newell
- Agency for Science, Technology and Research (A(∗)STAR), Singapore Immunology Network (SIgN), 138648 Singapore.
| |
Collapse
|
13
|
Roan F, Ziegler SF. Human Group 1 Innate Lymphocytes Are Negative for Surface CD3ε but Express CD5. Immunity 2018; 46:758-759. [PMID: 28514677 DOI: 10.1016/j.immuni.2017.04.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Florence Roan
- Immunology Program, Benaroya Research Institute, Seattle, Washington 98101, USA
| | - Steven F Ziegler
- Immunology Program, Benaroya Research Institute, Seattle, Washington 98101, USA; Department of Immunology, University of Washington School of Medicine, Seattle, Washington 98195, USA.
| |
Collapse
|
14
|
Simoni Y, Newell EW. Dissecting human ILC heterogeneity: more than just three subsets. Immunology 2017; 153:297-303. [PMID: 29140572 DOI: 10.1111/imm.12862] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/09/2017] [Accepted: 10/22/2017] [Indexed: 12/19/2022] Open
Abstract
Innate lymphoid cells (ILCs) have been divided into three distinct groups based on functional capacities, cytokine profiles and transcription factor expression. Studies performed mainly in mice have demonstrated the importance of ILCs in chronic inflammation, infection, allergy and cancer. In this review, we discuss the heterogeneity of human ILC and focus primarily on the taxonomy of human ILC cell subsets and their phenotypical and functional diversity. We summarize recent findings concerning the diversity of ILCs between and within the major subsets [natural killer (NK), ILC1, intra-epithelial ILC1 (ieILC1), ILC2, ILC3, lymphoid tissues inducer (LTi) and ILC progenitor (ILCP)], as well as the abundance of each in human tissues. We also discuss the similarities observed between groups of cells in term of receptors expressed and cytokines produced, and how these relate to the pleiotropic properties of each subset.
Collapse
Affiliation(s)
- Yannick Simoni
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
| | - Evan W Newell
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
| |
Collapse
|
15
|
Roan F, Stoklasek TA, Whalen E, Molitor JA, Bluestone JA, Buckner JH, Ziegler SF. CD4+ Group 1 Innate Lymphoid Cells (ILC) Form a Functionally Distinct ILC Subset That Is Increased in Systemic Sclerosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:2051-2062. [PMID: 26826243 PMCID: PMC4761490 DOI: 10.4049/jimmunol.1501491] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 01/01/2016] [Indexed: 12/16/2022]
Abstract
Innate lymphoid cells (ILC) are a heterogeneous group of cellular subsets that produce large amounts of T cell-associated cytokines in response to innate stimulation in the absence of Ag. In this study, we define distinct patterns of surface marker and cytokine expression among the ILC subsets that may further delineate their migration and function. Most notably, we found that the subset previously defined as group 1 ILC (ILC1) contains CD4(+) CD8(-), CD4(-) CD8(+), and CD4(-) CD8(-) populations. Although all ILC1 subsets shared characteristics with Th1 cells, CD4(+) ILC1 also demonstrated significant phenotypic and functional heterogeneity. We also show that the frequencies of CD4(+) ILC1 and NKp44(+) group 3 ILC, but not CD4(-) ILC1 or group 2 ILC, are increased in the peripheral blood of individuals with systemic sclerosis (SSc), a disease characterized by fibrotic and vascular pathology, as well as immune dysregulation. Furthermore, we demonstrate that CD4(+) and CD4(-) ILC1 are functionally divergent based on their IL-6Rα expression and that the frequency of IL-6Rα expression on ILC is altered in SSc. The distinct phenotypic and functional features of CD4(+) and CD4(-) ILC1 suggest that they may have differing roles in the pathogenesis of immune-mediated diseases, such as SSc.
Collapse
Affiliation(s)
- Florence Roan
- Benaroya Research Institute at Virginia Mason, Seattle, WA
- University of Washington, Division of Allergy and Infectious Diseases, Seattle, WA
| | | | | | - Jerry A. Molitor
- University of Minnesota, Division of Rheumatology, Minneapolis, MN
| | | | | | | |
Collapse
|
16
|
Fernandes MT, Dejardin E, dos Santos NR. Context-dependent roles for lymphotoxin-β receptor signaling in cancer development. Biochim Biophys Acta Rev Cancer 2016; 1865:204-19. [PMID: 26923876 DOI: 10.1016/j.bbcan.2016.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 02/03/2016] [Accepted: 02/24/2016] [Indexed: 12/20/2022]
Abstract
The LTα1β2 and LIGHT TNF superfamily cytokines exert pleiotropic physiological functions through the activation of their cognate lymphotoxin-β receptor (LTβR). Interestingly, since the discovery of these proteins, accumulating evidence has pinpointed a role for LTβR signaling in carcinogenesis. Early studies have shown a potential anti-tumoral role in a subset of solid cancers either by triggering apoptosis in malignant cells or by eliciting an anti-tumor immune response. However, more recent studies provided robust evidence that LTβR signaling is also involved in diverse cell-intrinsic and microenvironment-dependent pro-oncogenic mechanisms, affecting several solid and hematological malignancies. Consequently, the usefulness of LTβR signaling axis blockade has been investigated as a potential therapeutic approach for cancer. Considering the seemingly opposite roles of LTβR signaling in diverse cancer types and their key implications for therapy, we here extensively review the different mechanisms by which LTβR activation affects carcinogenesis, focusing on the diverse contexts and different models assessed.
Collapse
Affiliation(s)
- Mónica T Fernandes
- Centre for Biomedical Research (CBMR), University of Algarve, Faro 8005-139, Portugal; PhD Program in Biomedical Sciences, Department of Biomedical Sciences and Medicine, University of Algarve, Faro 8005-139, Portugal
| | - Emmanuel Dejardin
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Research, Molecular Biology of Diseases, University of Liège, Liège 4000, Belgium
| | - Nuno R dos Santos
- Centre for Biomedical Research (CBMR), University of Algarve, Faro 8005-139, Portugal; Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto 4200, Portugal; Institute of Pathology and Molecular Immunology, University of Porto (IPATIMUP), Porto 4200, Portugal.
| |
Collapse
|
17
|
Huang YF, Lo PC, Yen CL, Nigrovic PA, Chao WC, Wang WZ, Hsu GC, Tsai YS, Shieh CC. Redox Regulation of Pro-IL-1β Processing May Contribute to the Increased Severity of Serum-Induced Arthritis in NOX2-Deficient Mice. Antioxid Redox Signal 2015; 23:973-84. [PMID: 25867281 PMCID: PMC4624247 DOI: 10.1089/ars.2014.6136] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
AIMS To elucidate the role of reactive oxygen species (ROS) in arthritis and to identify targets of arthritis treatment in conditions with different levels of oxidant stress. RESULTS Through establishing an arthritis model by injecting arthritogenic serum into wild-type and NADPH oxidase 2 (NOX2)-deficient mice, we found that arthritis had a neutrophilic infiltrate and was more severe in Ncf1(-/-) mice, a mouse strain lacking the expression of the NCF1/p47(phox) component of NOX2. The levels of interleukin-1β (IL-1β) and IL-6 in inflamed joints were higher in Ncf1(-/-) than in controls. Antagonists of tumor necrosis factor-α (TNFα) and IL-1β were equally effective in suppressing arthritis in wild-type mice, while IL-1β blockade was more effective than TNFα blockade in Ncf1(-/-) mice. A treatment of caspase inhibitor and the combination treatment of a caspase inhibitor and a cathepsin inhibitor, but not a cathepsin inhibitor alone, suppressed arthritic severity in the wild-type mice, while a treatment of cathepsin inhibitor and the combination treatment of a caspase inhibitor and a cathepsin inhibitor, but not a caspase inhibitor alone, were effective in treating Ncf1(-/-) mice. Consistently, cathepsin B was found to proteolytically process pro-IL-1β to its active form and this activity was suppressed by ROS. INNOVATION This novel mechanism of a redox-mediated immune regulation of arthritis through leukocyte-produced ROS is important for devising an optimal treatment for patients with different levels of tissue ROS. CONCLUSION Our results suggest that ROS act as a negative feedback to constrain IL-1β-mediated inflammation, accounting for the more severe arthritis in the absence of NOX2.
Collapse
Affiliation(s)
- Ya-Fang Huang
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan
| | - Pei-Chi Lo
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan
| | - Chia-Liang Yen
- 2 Institute of Basic Medical Science, National Cheng Kung University College of Medicine , Tainan, Taiwan
| | - Peter Andrija Nigrovic
- 3 Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital , Boston, Massachusetts.,4 Division of Immunology, Boston Children's Hospital , Boston, Massachusetts
| | - Wen-Chen Chao
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan .,5 Department of Internal Medicine, Taichung Veteran General Hospital , Chiayi Branch, Chiayi, Taiwan
| | - Wei-Zhi Wang
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan
| | - George Chengkang Hsu
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan
| | - Yau-Sheng Tsai
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan
| | - Chi-Chang Shieh
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan .,6 Department of Pediatrics, National Cheng Kung University Hospital , Tainan, Taiwan
| |
Collapse
|
18
|
Characteristics of innate lymphoid cells (ILCs) and their role in immunological disorders (an update). Cell Immunol 2015; 298:66-76. [PMID: 26429626 DOI: 10.1016/j.cellimm.2015.09.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 09/04/2015] [Accepted: 09/15/2015] [Indexed: 02/05/2023]
Abstract
Innate lymphoid cells (ILCs) are a novel family of hematopoietic effectors and regulators of innate immunity. Although these cells are morphologically similar to B cells and T cells, however they do not express antigen receptors. ILCs seems to have emerging roles in innate immune responses against infectious or non-infectious microorganisms, protection of the epithelial barrier, lymphoid organogenesis and inflammation, tissue remodeling and regulating homeostasis of tissue stromal cells. In addition, it has recently been reported that ILCs have a crucial role in several disorders such as allergy and autoimmunity. Based on their phenotype and functions, ILCs are classified into three major groups called ILCs1, ILCs2, and ILCs3. Here we reviewed the most recent data concerning diverse ILC phenotypes, subclasses, functions in immune responses as well as in immune mediated disorders.
Collapse
|
19
|
Šedý J, Bekiaris V, Ware CF. Tumor necrosis factor superfamily in innate immunity and inflammation. Cold Spring Harb Perspect Biol 2014; 7:a016279. [PMID: 25524549 DOI: 10.1101/cshperspect.a016279] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The tumor necrosis factor superfamily (TNFSF) and its corresponding receptor superfamily (TNFRSF) form communication pathways required for developmental, homeostatic, and stimulus-responsive processes in vivo. Although this receptor-ligand system operates between many different cell types and organ systems, many of these proteins play specific roles in immune system function. The TNFSF and TNFRSF proteins lymphotoxins, LIGHT (homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for herpes virus entry mediator [HVEM], a receptor expressed by T lymphocytes), lymphotoxin-β receptor (LT-βR), and HVEM are used by embryonic and adult innate lymphocytes to promote the development and homeostasis of lymphoid organs. Lymphotoxin-expressing innate-acting B cells construct microenvironments in lymphoid organs that restrict pathogen spread and initiate interferon defenses. Recent results illustrate how the communication networks formed among these cytokines and the coreceptors B and T lymphocyte attenuator (BTLA) and CD160 both inhibit and activate innate lymphoid cells (ILCs), innate γδ T cells, and natural killer (NK) cells. Understanding the role of TNFSF/TNFRSF and interacting proteins in innate cells will likely reveal avenues for future therapeutics for human disease.
Collapse
Affiliation(s)
- John Šedý
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| | - Vasileios Bekiaris
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| | - Carl F Ware
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW T lymphocytes are critical to the pathogenesis of systemic rheumatic diseases. Understanding of the roles of T cells in disease has been enriched by the description of highly distinct effector subsets of CD4 T lymphocytes. The purpose of this review is to describe selected advances in the biology of T lymphocytes that are pertinent to the pathogenesis or treatment of rheumatic diseases. RECENT FINDINGS Knowledge is expanding about not only pathogenic effector T cell subsets, such as the TH17 cells, but also of regulatory T cells (Treg), the functions of which are defective, but correctable, in several rheumatic diseases. Although the initial agent that demonstrated a role for T cells in rheumatoid arthritis was CTLA4-Ig (abatacept), use of this biologic is now expanding to other rheumatic diseases. Moreover, effects of other biologics are now understood to in part be mediated by effects on T cell subsets. Experimental model systems in rodents continue to be valuable testing grounds for future approaches to treatment of human disease. Meanwhile, the roles of effector T cell subsets are becoming clearer in conditions such as Sjogren's syndrome and scleroderma. Finally, rheumatic diseases, including rheumatoid arthritis and spondyloarthropathies, have been critical for identification of new innate-like T cell subsets. SUMMARY Imbalances in the numbers and functions of specific T cell subsets are key pathogenic derangements in systemic rheumatic diseases, and these insights are leading to changes in clinical practice.
Collapse
|