• Title/Summary/Keyword: Tumor Immunity

Search Result 284, Processing Time 0.028 seconds

The Anti-tumor Activity of Vitamin C via the Increase of Fas (CD95) and MHC I expression on Human Stomach Cancer Cell Line, SNU1

  • Yu, Yeon-Sil;Bae, Se-Yeon;Kim, Hye-Min;Kim, Ye-Jin;Chu, Nag-Bum;Chu, Nag-Kyun;Kang, Jae-Seung;Lee, Wang-Jae
    • IMMUNE NETWORK
    • /
    • v.11 no.4
    • /
    • pp.210-215
    • /
    • 2011
  • It is already known that high concentration of vitamin C induces apoptosis on tumor cells. However, there is no report regarding the function of vitamin C on the modulation of immune susceptibility of cancer. Therefore, we investigated whether vitamin C can modulate immune susceptibility of tumor cells, especially on the induction of Fas-mediated apoptosis. First, the optimal concentration of vitamin C, which cannot induce damages on tumor cells for 36 hrs. We found that 2 mM of vitamin C did not show harmful effect. In addition, the optimal concentration of agonistic anti-Fas Abs for 18 hrs was examined. As a result, 400 ng/ml of agonistic anti-Fas Abs did not induce apoptosis on tumor cells. Next, we tried to find the effect of 2 mM of vitamin C on the modulation of the susceptibility to agonistic anti-Fas Abs. When tumor cells were cultured with 400 ng/ml of agonistic anti-Fas Abs for 18 hrs, after pre-treatment with 2 mM of vitamin C for 24 hrs, viability of cells was decreased. Interestingly, we found that the expression of Fas (CD95) and MHC class I was increased by the treatment of vitamin C. Taken together, vitamin C increases the susceptibility of tumor cells to anti-Fas Abs and the expression of Fas (CD95) and MHC class I on tumor cells.

The Effects of the Tumor Mass Size Inoculated in Immunologically Competent Balb/c Mice on Delayed-type Hypersensitivity Response (정상 면역 생쥐에 접종된 암세포주의 종괴 형성이 숙주의 지연성과민반응에 미치는 영향)

  • Lim, Hyun-Ja;Woo, A-Mi;Jung, Young-Ju;Kang, Jae-Seung;Shin, Dong-Hoon;Lee, Wang-Jae;Hwang, Young-Il
    • IMMUNE NETWORK
    • /
    • v.6 no.4
    • /
    • pp.185-191
    • /
    • 2006
  • Background: Based on outstanding progresses in animal experiments, vaccines for some human tumors have been developed. However, clinical effects of these vaccines have been far below than expected. This discrepancy might come from differences between animal models and human patients with respect to immunocompetency. The immune status of mice after tumor inoculation has not been well studied, which make us cautious in interpreting and applying the results from mice to human. We evaluated cell-mediated immune responses in mice after tumor cell inoculation. Methods: Mice were inoculated with TA3Ha, CT26, or 4T1. Delayed-type hypersensitivity (DTH) responses were induced 2-4 weeks after inoculation using 2,4-dinitro-1-fluorobenzene as an antigen. The relationships between the severity of DTH responses and the duration of tumor inoculation or the size of tumor mass were analyzed. Results: In T A3Ha groups, DTH response was elevated 2 weeks after inoculation, but depressed after 4 weeks, compared to the control group. When analyzed based on the sizes of tumor masses elicited, DTH responses were inversely related to the mass size, especially in those greater than 10 mm in diameter. In CT26 groups, while the duration after inoculation did not affect the severity of DTH responses, those with large mass showed depressed responses regardless the duration of inoculation. 4T1 cells grew so slowly that the size of tumor mass was small even 4 weeks after inoculation, and this group showed much higher DTH responses compared to that of tumor-free group. Conclusion: At least in an experimental setting where tumor model was induced by inoculating tumor cell lines into immunologically competent mice, the host immune response was elevated in early stage, and then depressed in late stage when the mass grew over a critical size.

A new aspect of an old friend: the beneficial effect of metformin on anti-tumor immunity

  • Kim, KyeongJin;Yang, Wen-Hao;Jung, Youn-Sang;Cha, Jong-ho
    • BMB Reports
    • /
    • v.53 no.10
    • /
    • pp.512-520
    • /
    • 2020
  • T-cell-based cancer immunotherapies, such as immune checkpoint blockers (ICBs) and chimeric antigen receptor (CAR)-T-cells, have significant anti-tumor effects against certain types of cancer, providing a new paradigm for cancer treatment. However, the activity of tumor infiltrating T-cells (TILs) can be effectively neutralized in the tumor microenvironment (TME) of most solid tumors, rich in various immunosuppressive factors and cells. Therefore, to improve the clinical outcomes of established T-cell-based immunotherapy, adjuvants that can comprehensively relieve multiple immunosuppressive mechanisms of TME are needed. In this regard, recent studies have revealed that metformin has several beneficial effects on anti-tumor immunity. In this mini-review, we understand the immunosuppressive properties of TME and how metformin comprehensively enhances anti-tumor immunity. Finally, we will discuss this old friend's potential as an adjuvant for cancer immunotherapy.

Identification of CM1 as a Pathogenic Factor in Inflammatory Diseases and Cancer

  • Bae, Se-Yeon;Kim, Hyem-In;Yu, Yeon-Sil;Lee, Na-Eun;Kong, Joo-Myoung;Kim, Hang-Rae;Hwang, Young-Il;Song, Yeong-Wook;Kang, Jae-Seung;Lee, Wang-Jae
    • IMMUNE NETWORK
    • /
    • v.11 no.3
    • /
    • pp.175-181
    • /
    • 2011
  • Background: CM1 (centrocyte/-blast marker 1) was defined by a mAb against concavabalin-A (ConA) activated PBMC. It is expressed in germinal center of human tonsil and on the surface of activated PBMC as well as cancer cells. Recently, increased productions of pro-inflammatory mediators were detected from activated PBMC by CM1 ligation. Methods: However, there is a limitation to explain the exact role of CM1 on inflammation and its related mechanisms, since the identity of CM1 is still not clarified. In our previous study, we have already confirmed that soluble form of CM1 was produced by Raji. Therefore, we performed Q-TOF analysis after immunoprecipitation of concentrated Raji culture supernatant using anti-CM1 mAbs. Results: As a result, we found that CM1 is identical to enolase-1(ENO1), a glycolytic enzyme, and we confirmed that results by silencing ENO1 using siRNA. It was also confirmed through competition assay between anti-CM1 and anti-ENO1 mAbs. Finally, we investigated the possible role of CM1 in inflammatory response and cancer. The ligation of CM1 on Raji cells with anti-CM1 mAbs induces the extensive production of prostaglandin $E_2(PGE_2)$. In addition, the increased activity of matrix metalloproteinase (MMP)-2/9 was shown in NCI-N87, stomach cancer cell line by CM1 stimulation. Conclusion: CM1 is identical to ENO1 and it might be an important role in the regulation of inflammatory responses.

The Effects of High-dose Vitamin C Administration on the Cell-mediated Immune Response in Mice (다용량 비타민 C 투여가 생쥐 세포매개면역반응에 미치는 영향)

  • Noh, Kahwa;Kim, Heun-gon;Shin, Young-ah;Lim, Hyunja;Mun, Sung-kyu;Lee, Yongtaek;Lee, Wang Jae;Lee, Dongsup;Hwang, Young-il
    • IMMUNE NETWORK
    • /
    • v.3 no.3
    • /
    • pp.211-218
    • /
    • 2003
  • Background: Vitamin C is an essential nutrient, taken as a daily supplement by many people. Recently, high-dose vitamin C is considered as a therapeutic regimen in some clinical situations. Until now, few studies have been done with the effects of high-dose vitamin C on the immune response. Methods: In this experiment, the effects of high-dose vitamin C on cell-mediated immune response in immunologically competent mice were evaluated. After intraperitoneal injection of 2.5, 5, or 10 mg/day of vitamin C for 10 days, delayed type hypersensitivity (DTH) was provoked against DNFB in the pinnae as a model for cell-mediated immune response. Severity of DTH reaction was evaluated as the thickness of pinnae, and the vitamin C levels were measured in the serum, liver, kidney, lung, pinnae, and splenocytes. Results: After challenge, the thickness increased at its peak on the $2^{nd}$ day in all groups. On the first day, the pinnae were thicker in the injected groups than in the control. On the contrary, the increment of the pinnae thickness was attenuated and the number of cells infiltrated in the site of DTH decreased proportionately to the amount of vitamin C administered from the second day on. With vitamin C exogenously given, the serum level peaked at 30 min after injection, and returned abruptly to its basal level without accumulation. However, it accumulated in the liver, kidney, and especially in the pinnae inflamed and splenopcytes, proportionately to the amount administered. Conclusion: Based on these results, it is suggested that, in one hand, exogenously administered high-dose vitamin C accumulated in the splenocytes and presumably changed the function of them resulting in the augmented cell-mediated immune response, as was revealed in the first day of DTH reaction. On the other hand, it seems likely that the vitamin C also showed anti-inflammatory effects.

Anti-tumor Immunity Induced by Tumor Cells Expressing a Membrane-bound Form of IL-2 and SDF-1

  • Choi, Jin-Wha;Lim, Ho-Yong;Chang, Mi-Ra;Cheon, Ji-Yeon;Kim, Young-Sang
    • Animal cells and systems
    • /
    • v.12 no.4
    • /
    • pp.193-201
    • /
    • 2008
  • The eventual goal of tumor immunotherapy is to develop a vaccine inducing a specific anti-tumor immunity. Cytokine gene therapy is an effective way at least in animal models, but limited efficacy and various side effects obstruct clinical applications. In this study, we developed a tumor vaccine expressing a membrane-bound form of IL-2(mbIL-2) and SDF-1 in B16F10 melanoma cells. The tumor clones expressing mbIL-2 showed reduced tumorigenicity, and additional expression of SDF-1 to mbIL-2 expressing tumor cells caused more severe reduction in tumorigenicity. However, expression of the SDF-1 alone did not affect on the tumorigenicity, probably because of limited production of SDF-1 in the SDF-1 transfected clones. When the mice once rejected mbIL-2/SDF-1 expressing tumor clone were re-challenged with wild type B16F10 tumor cells, all of the mice survived. This result suggests that mbIL-2/SDF-1 tumor clone is effective in inducing systemic anti-tumor immunity against wild type B16 melanoma. Furthermore, culture supernatant of tumor clones expressing SDF-1 induced lymphocyte migration in vitro. These results, all together, suggest that expression of mbIL-2 and SDF-1 in tumor cells enhances anti-tumor immune responses through different roles; the secreted SDF-1 may function as a chemoattractant to recruit immune cells to tumor vaccine injection site, and the mbIL-2 on tumor cells may provide costimulatory signal for CTL activation in physical contacts.

CD4+ cytotoxic T cells: an emerging effector arm of anti-tumor immunity

  • Seongmin Jeong;Nawon Jang;Minchae Kim;Il-Kyu Choi
    • BMB Reports
    • /
    • v.56 no.3
    • /
    • pp.140-144
    • /
    • 2023
  • While CD8+ cytotoxic T cells have long been considered the primary effector in controlling tumors, the involvement of CD4+ "helper" T cells in anti-tumor immunity has been underappreciated. The investigations of intra-tumoral T cells, fueled by the recent advances in genomic technologies, have led to a rethinking of the indirect role of CD4+ T cells that have traditionally been described as a "helper". Accumulating evidence from preclinical and clinical studies indicates that CD4+ T cells can acquire intrinsic cytotoxic properties and directly kill various types of tumor cells in a major histocompatibility complex class II (MHC-II)-dependent manner, as opposed to the indirect "helper" function, thus underscoring a potentially critical contribution of CD4+ cytotoxic T cells to immune responses against a wide range of tumor types. Here, we discuss the biological properties of anti-tumor CD4+ T cells with cytotoxic capability and highlight the emerging observations suggesting their more significant role in anti-tumor immunity than previously appreciated.

Induction of Enhancement of Anti-Tumor Immunity by Polysaccharides Fractionated from Acanthopanx Senticosus (가시오가피 다당체에 의한 항종양면역의 유도)

  • Yoon, Taek-Joon;Sung, Ji-Yeon;Yu, Kwang-Won;Lee, Ho;Lee, Kwang-Ho
    • Korean Journal of Pharmacognosy
    • /
    • v.38 no.2 s.149
    • /
    • pp.117-122
    • /
    • 2007
  • The specific activation of the immune system to control cancer growth in vivo has been a long-standing goal in cancer immunology. Whole tumor Iysates have been used either alone or combined with adjuvants to induce specific immune response in vivo. Here, we examined whether freezing/thawing (F/T) colon26-M3.1 tumor cell admixed with EN-3, glycoprotein purified from Acanthopanx Senticosus, could stimulate in vivo immunity by using a murine experimental tumor metastasis model produced by colon26-M3.1 carcinoma cells. Vaccination of mice with F/T treated colon26-M3.1 carcinoma cells in combination with EN-3 as an adjuvant resulted in a significant inhibition in tumor metastasis of mice against live colon26-M3.1 carcinoma challenge. In addition, the splenocytes from vaccinated mice exhibited a higher proliferating activity and secreted interferon-${\gamma}$. These results suggest that EN-3 can be applied to immunoadjuvant to enhance the antitumor immunity in vivo.

Anti-tumor Metastatic Effect and Activation of Innate Immunity by Extract of Mori Radicis Cortex (상백피(桑白皮)의 선천면역 활성화에 의한 항암 효과)

  • Jeong, Jae-Hyuk;Lee, Jin-Moo;Lee, Chang-Hoon;Cho, Jung-Hoon;Jang, Jun-Bock;Lee, Kyung-Sub
    • The Journal of Korean Obstetrics and Gynecology
    • /
    • v.22 no.1
    • /
    • pp.31-40
    • /
    • 2009
  • Purpose: This study was carried out to investigate the anti-tumor metastasis effect and activation of innate immunity by extracts of Mori radicis cortex. Methods: Anti-tumor metastatic experiment was conducted in vitro and in vivo by using colon 26-M3.1 carcinoma cell, L5178Y-R lymphoma cell and HeLa cell. To observe the activation of innate immunity by extracts of Mori radicis cortex, we estimated IL-6, IL-10, IL-12, TNF-${\alpha}$ from peritoneal macrophages. And we evaluated the activation of NK cell by using anti-asialo-GM1 serum. Results: We found that the administration of Mori radicis cortex extracts significantly inhibited tumor metastasis. In an in vitro cytotoxicity analysis, Mori radicis cortex affected tumor cell growth above specific concentration. Mori radicis cortex also stimulated peritoneal macrophage, which was followed by the production of various cytokines such as IL-6, IL-10, IL-12, TNF-${\alpha}$. The depletion of NK cells by anti-asialo GM1 serum partly abolished the inhibitory effect of Mori radicis cortex on tumor metastasis. Conclusion: Mori radicis cortex appears to have considerable activity on the anti-metastasis by activation of innate immunity.

CM1 Ligation Induces Apoptosis via Fas-FasL Interaction in Ramos Cells, but via Down-regulation of Bcl-2 and Subsequent Decrease of Mitochondrial Membrane Potential in Raji Cells

  • Lee, Young-Sun;Kim, Yeong-Seok;Kim, Dae-Jin;Hur, Dae-Young;Kang, Jae-Seung;Kim, Young-In;Hahm, Eun-Sil;Cho, Dae-Ho;Hwang, Young-Il;Lee, Wang-Jae
    • IMMUNE NETWORK
    • /
    • v.6 no.2
    • /
    • pp.59-66
    • /
    • 2006
  • Background: CM1 (Centrocyte/-blast Marker I) defined by a mAb developed against concanavalin-A activated PBMC, is expressed specifically on a subpopulation of centroblasts and centrocytes of human germinal center (GC) B cells. Burkitt lymphoma (BL) is a tumor consisting of tumor cells with the characteristics of GC B cell. Previously we reported that CM1 ligation with anti-CM1 mAb induced apoptosis in Ramos $(IgM^{high})$ and Raji $(IgM^{low})$ cells. Methods & Results: In the present study, we observed that CM1 ligation with anti-CM1 mAb induced Fas ligand and Fas expression in Ramos cells, but not in Raji cells. Furthermore, anti-Fas blocking antibody, ZB4, blocked CM1-mediated apoptosis effectively in Ramos cells, but not in Raji cells. Increased mitochondrial membrane permeabilization, which was measured by $DiOC_6$, was observed only in Raji cells. In contrast to no significant change of Bax known as pro-apoptotic protein, anti-apoptotic protein Bcl-2 was significantly decreased in Raji cells. In addition, we observed that CM1 ligation increased release of mitochondrial cytochrome c and upregulated caspase-9 activity in Raji cells. Conclusion: These results suggest that apoptosis induced by CM1-ligation is mediated by Fas-Fas ligand interaction in Ramos cells, whereas apoptosis is mediated by down-regulation of Bcl-2 and subsequent decrease of mitochondrial membrane potential in Raji cells.