1
|
Li D, Cao D, Sun Y, Cui Y, Zhang Y, Jiang J, Cao X. The roles of epigallocatechin gallate in the tumor microenvironment, metabolic reprogramming, and immunotherapy. Front Immunol 2024; 15:1331641. [PMID: 38348027 PMCID: PMC10859531 DOI: 10.3389/fimmu.2024.1331641] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/15/2024] [Indexed: 02/15/2024] Open
Abstract
Cancer, a disease that modern medicine has not fully understood and conquered, with its high incidence and mortality, deprives countless patients of health and even life. According to global cancer statistics, there were an estimated 19.3 million new cancer cases and nearly 10 million cancer deaths in 2020, with the age-standardized incidence and mortality rates of 201.0 and 100.7 per 100,000, respectively. Although remarkable advancements have been made in therapeutic strategies recently, the overall prognosis of cancer patients remains not optimistic. Consequently, there are still many severe challenges to be faced and difficult problems to be solved in cancer therapy today. Epigallocatechin gallate (EGCG), a natural polyphenol extracted from tea leaves, has received much attention for its antitumor effects. Accumulating investigations have confirmed that EGCG can inhibit tumorigenesis and progression by triggering apoptosis, suppressing proliferation, invasion, and migration, altering tumor epigenetic modification, and overcoming chemotherapy resistance. Nevertheless, its regulatory roles and biomolecular mechanisms in the immune microenvironment, metabolic microenvironment, and immunotherapy remain obscure. In this article, we summarized the most recent updates about the effects of EGCG on tumor microenvironment (TME), metabolic reprogramming, and anti-cancer immunotherapy. The results demonstrated EGCG can promote the anti-cancer immune response of cytotoxic lymphocytes and dendritic cells (DCs), attenuate the immunosuppression of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs), and inhibit the tumor-promoting functions of tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), and various stromal cells including cancer-associated fibroblasts (CAFs), endothelial cells (ECs), stellate cells, and mesenchymal stem/stromal cells (MSCs). Additionally, EGCG can suppress multiple metabolic reprogramming pathways, including glucose uptake, aerobic glycolysis, glutamine metabolism, fatty acid anabolism, and nucleotide synthesis. Finally, EGCG, as an immunomodulator and immune checkpoint blockade, can enhance immunotherapeutic efficacy and may be a promising candidate for antitumor immunotherapy. In conclusion, EGCG plays versatile regulatory roles in TME and metabolic reprogramming, which provides novel insights and combined therapeutic strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Dongming Li
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Donghui Cao
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Yuanlin Sun
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yingnan Cui
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yangyu Zhang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Jing Jiang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Xueyuan Cao
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
2
|
Chen X, Xu Z, Lu M, Ding W, Zhong J, Deng S, Li S, Miao J, Liu X, Wen Q, Ye S, Li C, Li H. Paeonol inhibits melanoma growth by targeting PD1 through upregulation of miR-139-5p. Biochem Biophys Res Commun 2023; 656:86-96. [PMID: 36958259 DOI: 10.1016/j.bbrc.2023.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 02/26/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
The abnormal immune response mediated by malignant melanoma is related to PD1. Paeonol has pharmacological antitumor activity. Previous studies have indicated that paeonol induces tumor cell apoptosis, but its underlying mechanism in tumor immunity remains unknown. In this study, malignant melanoma was established in normal and thymectomized mice to determine the important role of the thymus in the antitumor effects of paeonol. Paeonol-treated thymocytes were cocultured with melanoma cell spheres to further evaluate the regulatory role of thymocytes in tumor immune dysfunction. Studies have shown that PD1 may be targeted by miR-139-5p. Our results revealed that tumor-induced thymic atrophy was significantly accompanied by high PD1 expression and low miR-139-5p expression. Interestingly, paeonol significantly reversed thymic atrophy and largely protected thymocytes against low PD1 expression and high miR-139-5p expression. Dual-luciferase assays indicated that miR-139-5p interacted with the 3' untranslated region (3'-UTR) of PD1. These results showed that paeonol alleviates PD1-mediated antitumor immunity by reducing miR-139-5p expression and demonstrated a novel mechanism for melanoma immunotherapy.
Collapse
Affiliation(s)
- Xianjie Chen
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China; Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Zhenyuan Xu
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China; Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Meng Lu
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China; Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Wenjun Ding
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China; Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Jun Zhong
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China; Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Suihui Deng
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China; Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Siyan Li
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China; Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Jifei Miao
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, PR China
| | - Xiaoyi Liu
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China; Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Quan Wen
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Sen Ye
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China; Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China.
| | - Chun Li
- School of Nursing Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China.
| | - Hui Li
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China; Department of Human Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China.
| |
Collapse
|
3
|
Bednarz-Misa I, Bromke MA, Krzystek-Korpacka M. Interleukin (IL)-7 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1290:9-49. [PMID: 33559853 DOI: 10.1007/978-3-030-55617-4_2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Interleukin (IL)-7 plays an important immunoregulatory role in different types of cells. Therefore, it attracts researcher's attention, but despite the fact, many aspects of its modulatory action, as well as other functionalities, are still poorly understood. The review summarizes current knowledge on the interleukin-7 and its signaling cascade in context of cancer development. Moreover, it provides a cancer-type focused description of the involvement of IL-7 in solid tumors, as well as hematological malignancies.The interleukin has been discovered as a growth factor crucial for the early lymphocyte development and supporting the growth of malignant cells in certain leukemias and lymphomas. Therefore, its targeting has been explored as a treatment modality in hematological malignancies, while the unique ability to expand lymphocyte populations selectively and without hyperinflammation has been used in experimental immunotherapies in patients with lymphopenia. Ever since the early research demonstrated a reduced growth of solid tumors in the presence of IL-7, the interleukin application in boosting up the anticancer immunity has been investigated. However, a growing body of evidence indicative of IL-7 upregulation in carcinomas, facilitating tumor growth and metastasis and aiding drug-resistance, is accumulating. It therefore becomes increasingly apparent that the response to the IL-7 stimulus strongly depends on cell type, their developmental stage, and microenvironmental context. The interleukin exerts its regulatory action mainly through phosphorylation events in JAK/STAT and PI3K/Akt pathways, while the significance of MAPK pathway seems to be limited to solid tumors. Given the unwavering interest in IL-7 application in immunotherapy, a better understanding of interleukin role, source in tumor microenvironment, and signaling pathways, as well as the identification of cells that are likely to respond should be a research priority.
Collapse
Affiliation(s)
- Iwona Bednarz-Misa
- Department of Medical Biochemistry, Wroclaw Medical University, Wroclaw, Poland
| | - Mariusz A Bromke
- Department of Medical Biochemistry, Wroclaw Medical University, Wroclaw, Poland
| | | |
Collapse
|
4
|
Feitosa IB, Mori B, Teles CBG, Costa AGD. What are the immune responses during the growth of Ehrlich's tumor in ascitic and solid form? Life Sci 2020; 264:118578. [PMID: 33058910 DOI: 10.1016/j.lfs.2020.118578] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/25/2020] [Accepted: 10/03/2020] [Indexed: 10/23/2022]
Abstract
Traditionally, Ehrlich's tumor is used in experimental oncology to investigate the therapeutic capacity of different synthetic chemotherapeutic agents or to evaluate the antitumoral activity of different substances of natural origin. However, the understanding of immune mechanisms during Ehrlich carcinogenesis is still limited. In this review, we seek to describe the immune response during Ehrlich's tumor growth, and natural response without the influence of pharmacological administration, immunotherapies or concomitant challenges. The study followed the recommendations of the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA). A systematic review was carried out that included experimental trials with mice challenged with Ehrlich's tumor. The research was carried out in three databases including MEDLINE/PubMed, Scopus, Latin American and Caribbean Literature in Health Sciences (LILACS). The searches resulted in 913 papers being found, of which 55 articles were considered eligible, and of these 55, 29 were selected for analysis. Findings indicate that there is an increase in the expression of M2 and T Helper (TH2) macrophages and of the cytokines IL-17, IL-1B, IL-6 and PGE in the ascitic form of Ehrlich. These phenotypic expressions are also found in ascitic neoplasms in humans. Ehrlich's solid tumor was characterized by increased expression of CD4, CD8, neutrophils and TNF-a, Foxp3 + and Qa-2 +, and these characteristics are analogous to human breasts cancers. It is our understanding that further studies are needed to assess the immune mechanisms in Ehrlich's tumor, since these findings can be used to improve cancer treatments that are analogous to Ehrlich's tumor.
Collapse
Affiliation(s)
- Ivan Brito Feitosa
- Instituto de Ciências Biológicas, Programa de Pós-graduação em Imunologia Básica e Aplicada, PPGIBA, Universidade Federal do Amazonas (UFAM), Manaus, Amazonas State, Brazil.
| | - Bruno Mori
- Instituto de Ciências Biológicas, Programa de Pós-graduação em Imunologia Básica e Aplicada, PPGIBA, Universidade Federal do Amazonas (UFAM), Manaus, Amazonas State, Brazil
| | - Carolina Bioni Garcia Teles
- Plataforma Técnica de Bioensaio de Malária e Leishmaniose, Fundação Oswaldo Cruz, Fiocruz, Rondônia/Centro Universitário São Lucas, Departamento de Medicina/Instituto Nacional de Epidemiologia na Amazônia Ocidental, Porto Velho, Rondônia State, Brazil
| | - Alysson Guimarães da Costa
- Instituto de Ciências Biológicas, Programa de Pós-graduação em Imunologia Básica e Aplicada, PPGIBA, Universidade Federal do Amazonas (UFAM), Manaus, Amazonas State, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas, Manaus, Amazonas State, Brazil
| |
Collapse
|
5
|
Sun S, Ji H, Feng Y, Kang Y, Yu J, Liu A. A novel mechanism of tumor-induced thymic atrophy in mice bearing H22 hepatocellular carcinoma. Cancer Manag Res 2018; 10:417-424. [PMID: 29551914 PMCID: PMC5842769 DOI: 10.2147/cmar.s157512] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Background Thymic atrophy was discovered in tumor-bearing mice in recent years. Methods Flow cytometry was carried out including Annexin V-FITC/PI double staining, PI staining, Terminal dUTP nick-end labeling, CD3-FITC/CD19-PE and CD8-FITC/CD4-PE double staining. Enzyme-linked immunosorbent assay and polymerase chain reaction were also investigated. Results According to our experiments, we demonstrated that no signs of apoptosis in thymocytes were found in H22-bearing mice, while the proportions of CD4+ T cells and CD8+ T cells in thymuses were remarkably increased, the opposite tendency was found in peripheral bloods, and only CD3+CD8+ T cells were discovered in H22 solid tumors. We further discovered that the level of thymosin alpha 1 (Tα1) and the expression of Wnt4 in thymus of H22-bearing mice were significantly improved than control, which indicated the active proliferation and differentiation of thymocytes. Our study revealed that CD8+ T cells could not effectively eliminate H22 cells independently when CD4+ T cells were suppressed by tumors, while the body would only enhance the differentiation and maturation of T cells in thymuses and release them to solid tumor to reinforce antitumor immunocompetence, leading to a vicious cycle which finally led to thymic atrophy. Conclusion Our data propose a novel mechanism of tumor-induced thymic atrophy regulated by abnormal immunoreaction and may provide new ideas for the immunotherapy of tumors.
Collapse
Affiliation(s)
- Sujun Sun
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Haiyu Ji
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Yingying Feng
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Yu Kang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Juan Yu
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Anjun Liu
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| |
Collapse
|
6
|
Bhattacharyya S, Saha J. Tumour, Oxidative Stress and Host T Cell Response: Cementing the Dominance. Scand J Immunol 2016; 82:477-88. [PMID: 26286126 DOI: 10.1111/sji.12350] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/10/2015] [Indexed: 12/23/2022]
Abstract
Reactive oxygen species (ROS) and free radicals are produced intrinsically during normal cellular metabolic processes or extrinsically due to ionizing radiations, UV rays, xenobiotic insult, etc. ROS are important signal mediators and are used by the immune system to destroy pathogens, but as these are highly reactive, they also have the capacity to cause DNA damage and alter protein and lipid components of a cell. As a result, cells have evolved a tight regulation of internal redox environment that involves a balanced interplay between free radicals produced and quenched by cellular antioxidants and enzyme systems. Any deregulation of this subtle balance can result in oxidative stress that can lead to various pathological conditions including cancer. Oxidative stress can be a cause of neoplasia, or it can be induced by a growing tumour itself. The link existing between oxidative stress and inflammation is also very strong. Suppressed cellular immune system, especially effector T cell system, is a characteristic of tumour-bearing host. Both the direct oxidative stress caused by tumour cell(s) and oxidative stress mediators present in tumour microenvironment play a significant role in the suppression of effector T cell function and induction of T cell death. This review discusses in detail the complex interplay between tumour-stroma-immune system in the light of oxidative stress that dominates every phase of cancer including initiation, progression and establishment. This review also addresses in detail the mechanisms of oxidative stress-induced T cell dysfunction in tumour-bearing host and also briefly points out the possible therapeutic interventions.
Collapse
Affiliation(s)
- S Bhattacharyya
- Department of Zoology, Sidho Kanho Birsha University, Purulia, West Bengal, India
| | - J Saha
- Department of Zoology, Sidho Kanho Birsha University, Purulia, West Bengal, India
| |
Collapse
|
7
|
Song Y, Yu R, Wang C, Chi F, Guo Z, Zhu X. Disruption of the Thymic Microenvironment Is Associated with Thymic Involution of Transitional Cell Cancer. Urol Int 2014; 92:104-15. [DOI: 10.1159/000353350] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 05/29/2013] [Indexed: 11/19/2022]
|
8
|
Saha S, Hossain DMS, Mukherjee S, Mohanty S, Mazumdar M, Mukherjee S, Ghosh UK, Nayek C, Raveendar C, Khurana A, Chakrabarty R, Sa G, Das T. Calcarea carbonica induces apoptosis in cancer cells in p53-dependent manner via an immuno-modulatory circuit. Altern Ther Health Med 2013; 13:230. [PMID: 24053127 PMCID: PMC3856502 DOI: 10.1186/1472-6882-13-230] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 09/12/2013] [Indexed: 11/10/2022]
Abstract
BACKGROUND Complementary medicines, including homeopathy, are used by many patients with cancer, usually alongside with conventional treatment. However, the molecular mechanisms underneath the anti-cancer effect, if any, of these medicines have still remained unexplored. To this end we attempted to evaluate the efficacy of calcarea carbonica, a homeopathic medicine, as an anti-cancer agent and to delineate the detail molecular mechanism(s) underlying calcerea carbonica-induced tumor regression. METHODS To investigate and delineate the underlying mechanisms of calcarea carbonica-induced tumor regression, Trypan blue dye-exclusion test, flow cytometric, Western blot and reverse transcriptase-PCR techniques were employed. Further, siRNA transfections and inhibitor studies were used to validate the involvement of p53 pathway in calcarea carbonica-induced apoptosis in cancer cells. RESULTS Interestingly, although calcarea carbonica administration to Ehrlich's ascites carcinoma (EAC)- and Sarcoma-180 (S-180)-bearing Swiss albino mice resulted in 30-35% tumor cell apoptosis, it failed to induce any significant cell death in ex vivo conditions. These results prompted us to examine whether calcarea carbonica employs the immuno-modulatory circuit in asserting its anti-tumor effects. Calcarea carbonica prevented tumor-induced loss of effector T cell repertoire, reversed type-2 cytokine bias and attenuated tumor-induced inhibition of T cell proliferation in tumor-bearing host. To confirm the role of immune system in calcarea carbonica-induced cancer cell death, a battery of cancer cells were co-cultured with calcarea carbonica-primed T cells. Our results indicated a "two-step" mechanism of the induction of apoptosis in tumor cells by calcarea carbonica i.e., (1) activation of the immune system of the host; and (2) induction of cancer cell apoptosis via immuno-modulatory circuit in p53-dependent manner by down-regulating Bcl-2:Bax ratio. Bax up-regulation resulted in mitochondrial transmembrane potential loss and cytochrome c release followed by activation of caspase cascade. Knocking out of p53 by RNA-interference inhibited calcarea carbonica-induced apoptosis thereby confirming the contribution of p53. CONCLUSION These observations delineate the significance of immuno-modulatory circuit during calcarea carbonica-mediated tumor apoptosis. The molecular mechanism identified may serve as a platform for involving calcarea carbonica into immunotherapeutic strategies for effective tumor regression.
Collapse
|
9
|
Ghosh S, Adhikary A, Chakraborty S, Nandi P, Mohanty S, Chakraborty S, Bhattacharjee P, Mukherjee S, Putatunda S, Chakraborty S, Chakraborty A, Sa G, Das T, Sen PC. Nifetepimine, a dihydropyrimidone, ensures CD4+ T cell survival in a tumor microenvironment by maneuvering sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA). J Biol Chem 2012; 287:32881-96. [PMID: 22851172 DOI: 10.1074/jbc.m112.357889] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Multiple mechanisms have been proposed by which tumors induce T cell apoptosis to circumvent tumor immune-surveillance. Although sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA) have long been known to regulate intracellular Ca(2+) homeostasis, few studies have examined the role of SERCA in processes of T lymphocyte survival and activation. In this context it remains largely unexplored as to how tumors jeopardize SERCA function to disable T cell-mediated anti-tumor immunity. Here, we show that human CD4(+) T cells in the presence of tumor conditions manifested an up-regulation of SERCA3 expression that resulted in development of endoplasmic reticulum stress leading to CD4(+) T cell apoptosis. Prostaglandin E(2) produced by the tumor cell plays a critical role in up-regulating SERCA3 by enhancing the binding of its transcription factor Sp1. Gene manipulation and pharmacological approaches further established that an increase in SERCA expression also resulted in subsequent inhibition of PKCα and -θ and retention of NFκB in the cytosol; however, down-modulation of SERCA3 expression by a dihydropyrimidone derivative, ethyl-4-(3-nitro)-phenyl-6-methyl-2-oxo-1,2,3,4-tetrahydropyrimidine-5 carboxylate (nifetepimine), protected the CD4(+) T cells from tumor-induced apoptosis. In fact, nifetepimine-mediated restoration of PKC activity resulted in nuclear translocation of p65NFκB, thereby ensuring its survival. Studies further undertaken in a tumor-bearing mice model revalidated the immunoprotective role of nifetepimine. Our present study thus strongly suggests that imbalance in cellular calcium homeostasis is an important factor leading to CD4(+) T cell death during cancer and holds promise that nifetepimine may have the potential to be used as an immunorestoring agent in cancer bearers.
Collapse
Affiliation(s)
- Swatilekha Ghosh
- Division of Molecular Medicine, Bose Institute, P1/12 Calcutta Improvement Trust Scheme VIIM Kolkata 700054, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Zhao R, Chen Z, Jia G, Li J, Cai Y, Shao X. Protective effects of diosmetin extracted from Galium verum L. on the thymus of U14-bearing mice. Can J Physiol Pharmacol 2011; 89:665-73. [DOI: 10.1139/y11-058] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Diosmetin (DGVL) extracted from the traditional Chinese herb Galium verum L. has been found to have anticancer activity. In this study, the effects of DGVL on the thymus of U14-bearing mice were investigated. Using flow cytometry, peripheral blood lymphocytes were characterized based on the expression of surface markers for T helper cells (CD4+) and T suppressor cells (CD8+). Serum levels of tumor necrosis factor α (TNF-α), interleukin-2 (IL-2), IL-10, and transforming growth factor β1 (TGF-β1) and a cell proliferation assay were determined with an enzyme-linked immunosorbent assay. The expression of Fas and Fas ligand (FasL) on the thymus was determined by Western blotting. Our results showed that DGVL inhibited tumor growth and significantly increased the thymus weight compared with the control. Also, DGVL elevated serum levels of IL-2 and significantly reduced levels of TNF-α, TGF-β1, and IL-10 in a dose-dependent manner. Histological study and terminal dUTP nick end labeling staining results showed that DGVL protected thymus tissue against the onslaught of tumor growth by inhibiting thymus lymphocyte apoptosis. The cell proliferation assay revealed that DGVL might promote more thymus lymphocytes towards proliferation. Furthermore, the ratio of CD4+/CD8+ T lymphocytes was significantly increased from 0.69 to 2.29 by treatment with DGVL. Immunoblotting analyses revealed that the expression of Fas and FasL on the thymus was lower in mice in the DGVL treatment group than in the control mice. In conclusion, DGVL can inhibit tumor growth and protect tumor-induced apoptosis of the thymus, and the mechanism is closely associated with reduced cell death in the thymus and a Fas–FasL-dependent pathway.
Collapse
Affiliation(s)
- Rui Zhao
- Department of Pharmaceutical Engineering, College of Life Science and Biotechnology, Heilongjiang August First Land Reclamation University, Daqing High-Tech Industrial Development Zone, 163319, P.R. China
| | - Zhibao Chen
- Department of Pharmaceutical Engineering, College of Life Science and Biotechnology, Heilongjiang August First Land Reclamation University, Daqing High-Tech Industrial Development Zone, 163319, P.R. China
| | - Guiyan Jia
- Department of Pharmaceutical Engineering, College of Life Science and Biotechnology, Heilongjiang August First Land Reclamation University, Daqing High-Tech Industrial Development Zone, 163319, P.R. China
| | - Jian Li
- Department of Biological Engineering, School of Environment and Chemistry Engineering, Yanshan University, 438 Hebei Street, Qinhuangdao 066004, P.R. China
| | - Yaping Cai
- Department of Pharmaceutical Engineering, College of Life Science and Biotechnology, Heilongjiang August First Land Reclamation University, Daqing High-Tech Industrial Development Zone, 163319, P.R. China
| | - Xingyue Shao
- Department of Gynaecology and Obstetrics, Daqing Oilfield Hospital, Daqing 163311, P.R China
| |
Collapse
|
11
|
Ikehara S. Thymus transplantation for treatment of cancer: lessons from murine models. Expert Rev Clin Immunol 2011; 7:205-11. [PMID: 21426258 DOI: 10.1586/eci.10.100] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
It is well known that tumor-bearing mammals, including humans, show decreased T-cell function due to involution of the thymus. This decrease results in faster tumor growth, susceptibility to infection and reduced life expectancy. Thus, the best strategy to restore T-cell function might be to transplant the thymus from the fetus or newborn. Based on this hypothesis, this article introduces our recent findings using mice and evidence is provided that, in humans, thymus transplantation in conjunction with bone marrow transplantation could become a valuable strategy for suppressing tumor growth, thereby prolonging survival.
Collapse
Affiliation(s)
- Susumu Ikehara
- Department of Stem Cell Disorders, Kansai Medical University, Osaka, Japan.
| |
Collapse
|
12
|
Hosaka N, Cui W, Zhang Y, Takaki T, Inaba M, Ikehara S. Prolonged survival in mice with advanced tumors treated with syngeneic or allogeneic intra-bone marrow-bone marrow transplantation plus fetal thymus transplantation. Cancer Immunol Immunother 2010; 59:1121-30. [PMID: 20229083 PMCID: PMC11030866 DOI: 10.1007/s00262-010-0840-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2009] [Accepted: 02/23/2010] [Indexed: 12/20/2022]
Abstract
Thymic function decreases in line with tumor progression in patients with cancer, resulting in immunodeficiency and a poor prognosis. In the present study, we attempted to restore thymic function by BALB/c (H-2(d)) syngeneic (Syn), or B6 (H-2(b)) allogeneic (Allo) bone marrow transplantation (BMT) using intra-bone marrow-bone marrow transplantation (IBM-BMT) plus Syn-, Allo- or C3H (H-2(k)) 3rd-party fetal thymus transplantation (TT). Although the BALB/c mice with advanced tumors (Meth-A sarcoma; H-2(d), >4 cm(2)) treated with either Syn- or Allo-BMT alone showed a slight improvement in survival compared with non-treated controls, the mice treated with BMT + TT showed a longer survival. The mice treated with Allo-BMT + Allo-TT or 3rd-party TT showed the longest survival. Interestingly, although there was no difference in main tumor size among the BMT groups, lung metastasis was significantly inhibited by Allo-BMT + Allo-TT or 3rd-party TT. Numbers of CD4(+) and CD8(+) T cells, Con A response, and IFN-gamma production increased significantly, whereas number of Gr-1(+)/CD11b(+) myeloid suppressor cells and the percentage of FoxP3(+) cells in CD4(+) T cells significantly decreased in these mice. Furthermore, there was a positive correlation between survival days and the number of T cells or T cell function, while there was a negative correlation between survival days and lung metastasis, the number of Gr-1(+)/CD11b(+) cells, or the percentage of FoxP3(+) cells. These results suggest that BMT + TT, particularly Allo-BMT + Allo-TT or 3rd-party TT, is most effective in prolonging survival as a result of the restoration of T cell function in hosts with advanced tumors.
Collapse
MESH Headings
- Animals
- Bone Marrow Transplantation/methods
- CD11b Antigen/immunology
- Cell Line, Tumor
- Cytokines/metabolism
- Female
- Fetal Tissue Transplantation/methods
- Flow Cytometry
- Forkhead Transcription Factors/metabolism
- Interferon-gamma/metabolism
- Lung Neoplasms/immunology
- Lung Neoplasms/secondary
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/surgery
- Receptors, Chemokine/immunology
- Survival Analysis
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Thymus Gland/embryology
- Thymus Gland/transplantation
- Time Factors
- Transplantation, Homologous
- Transplantation, Isogeneic
Collapse
Affiliation(s)
- Naoki Hosaka
- First Department of Pathology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8506 Japan
| | - Wenhao Cui
- First Department of Pathology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8506 Japan
| | - Yuming Zhang
- First Department of Pathology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8506 Japan
- Department of Pediatrics, Nanfang Hospital, Guangzhou, 510515 China
| | - Takashi Takaki
- First Department of Pathology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8506 Japan
| | - Muneo Inaba
- First Department of Pathology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8506 Japan
| | - Susumu Ikehara
- First Department of Pathology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi, Osaka 570-8506 Japan
| |
Collapse
|
13
|
Li J, Li Q, Peng Y, Zhao R, Han Z, Gao D. Protective effects of fraction 1a of polysaccharides isolated from Solanum nigrum Linne on thymus in tumor-bearing mice. JOURNAL OF ETHNOPHARMACOLOGY 2010; 129:350-356. [PMID: 20371278 DOI: 10.1016/j.jep.2010.03.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 02/09/2010] [Accepted: 03/27/2010] [Indexed: 05/29/2023]
Abstract
AIM OF THE STUDY To further screen out the anti-tumor active compound of polysaccharides isolated from Solanum nigrum Linne (SNL-P), which had shown to have anti-cervical cancer and modulating properties, and evaluate the thymus protective effects of this active compound. MATERIAL AND METHODS SNL-P was separated and purified by column chromatography, and its anti-cervical cancer activity was evaluated by mice models injected of ascites U14 cells. Furthermore, the protective effect of fraction 1a of SNL-P (SNL-P1a) on the thymus tissue of tumor-bearing mice was evaluated by histological study and TUNEL staining. Finally, the protein expression of Bcl-2 and Bax gene were assayed by immunohistochemistry. RESULTS SNL-P1a has shown a marked inhibition effect on U14 cevical cancer, it restore the ratio of CD4(+)/CD8(+) peripheral blood T-lymphocyte subpopulation. Histological study and TUNEL staining results showed that SNL-P1a protect thymus tissue against the onslaught of tumor by inhibiting thymus lymphocyte apoptosis, and immunohistochemistry assay displayed that SNL-P1a treatment could increase Bcl-2/Bax ratio in thymus lymphocytes of tumor-bearer, which might promote more thymus lymphocytes towards proliferation. CONCLUSION SNL-P1a had significant growth inhibition effect on U14 cervical cancer and protective effect on thymus tissue of tumor-bearing mice.
Collapse
Affiliation(s)
- Jian Li
- College of Environmental & Chemical Engineering, Yanshan University, Qinhuangdao, Hebei Province, China
| | | | | | | | | | | |
Collapse
|
14
|
Chattopadhyay S, Bhattacharyya S, Saha B, Chakraborty J, Mohanty S, Sakib Hossain DM, Banerjee S, Das K, Sa G, Das T. Tumor-shed PGE(2) impairs IL2Rgammac-signaling to inhibit CD4 T cell survival: regulation by theaflavins. PLoS One 2009; 4:e7382. [PMID: 19812686 PMCID: PMC2753647 DOI: 10.1371/journal.pone.0007382] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2009] [Accepted: 08/28/2009] [Indexed: 01/18/2023] Open
Abstract
Background Many tumors are associated with decreased cellular immunity and elevated levels of prostaglandin E2 (PGE2), a known inhibitor of CD4+ T cell activation and inducer of type-2 cytokine bias. However, the role of this immunomodulator in the survival of T helper cells remained unclear. Since CD4+ T cells play critical roles in cell-mediated immunity, detail knowledge of the effect tumor-derived PGE2 might have on CD4+ T cell survival and the underlying mechanism may, therefore, help to overcome the overall immune deviation in cancer. Methodology/Principal Findings By culturing purified human peripheral CD4+ T cells or Jurkat cells with spent media of theaflavin- or celecoxib-pre-treated MCF-7 cells, we show that tumor-shed PGE2 severely impairs interleukin 2 receptor γc (IL2Rγc)-mediated survival signaling in CD4+ T cells. Indeed, tumor-shed PGE2 down-regulates IL2Rγc expression, reduces phosphorylation as well as activation of Janus kinase 3 (Jak-3)/signal transducer and activator of transcription 5 (Stat-5) and decreases Bcl-2/Bax ratio thereby leading to activation of intrinsic apoptotic pathway. Constitutively active Stat-5A (Stat-5A1*6) over-expression efficiently elevates Bcl-2 levels in CD4+ T cells and protects them from tumor-induced death while dominant-negative Stat-5A over-expression fails to do so, indicating the importance of Stat-5A-signaling in CD4+ T cell survival. Further support towards the involvement of PGE2 comes from the results that (a) purified synthetic PGE2 induces CD4+ T cell apoptosis, and (b) when knocked out by small interfering RNA, cyclooxygenase-2 (Cox-2)-defective tumor cells fail to initiate death. Interestingly, the entire phenomena could be reverted back by theaflavins that restore cytokine-dependent IL2Rγc/Jak-3/Stat-5A signaling in CD4+ T cells thereby protecting them from tumor-shed PGE2-induced apoptosis. Conclusions/Significance These data strongly suggest that tumor-shed PGE2 is an important factor leading to CD4+ T cell apoptosis during cancer and raise the possibility that theaflavins may have the potential as an effective immunorestorer in cancer-bearer.
Collapse
Affiliation(s)
- Sreya Chattopadhyay
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata, India
| | - Sankar Bhattacharyya
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata, India
| | - Baisakhi Saha
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata, India
| | - Juni Chakraborty
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata, India
| | - Suchismita Mohanty
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata, India
| | | | - Shuvomoy Banerjee
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata, India
| | - Kaushik Das
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata, India
| | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata, India
| | - Tanya Das
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata, India
- * E-mail:
| |
Collapse
|
15
|
Letchoumy PV, Subapriya R, Nagini S, Abraham SK. Protective Effect of Black Tea Polyphenols Against 7,12-Dimethylbenz[A]Anthracene-Induced Genotoxicity and Oxidative Stress During Hamster Buccal Pouch Carcinogenesis. Toxicol Mech Methods 2008; 17:93-100. [DOI: 10.1080/15376510600860193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|