Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleExperimental Studies
Open Access

Dimethyl Fumarate Induces Apoptosis via Inhibiting NF-κB and STAT3 Signaling in Adult T-cell Leukemia/Lymphoma Cells

TAKAHIRO MAETA, TSUYOSHI SATO, KAZUYA ASANO and SHIGEKI ITO
Anticancer Research May 2022, 42 (5) 2301-2309; DOI: https://doi.org/10.21873/anticanres.15709
TAKAHIRO MAETA
Division of Hematology & Oncology, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
TSUYOSHI SATO
Division of Hematology & Oncology, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KAZUYA ASANO
Division of Hematology & Oncology, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SHIGEKI ITO
Division of Hematology & Oncology, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: shigei@iwate-med.ac.jp
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Adult T-cell leukemia (ATL) is a peripheral T lymphocytic malignancy caused by human T-cell leukemia virus type 1 (HTLV-1) infection. Despite treatment that includes novel agents that have been developed, most of ATL patients relapse and acquire multidrug resistance. As a result, the creation of newer agents is critical. Dimethyl fumarate (DMF) has several effects in cancer cells, including cell signaling, proliferation and cell death. However, its antitumor effects on ATL cells remain unknown. In this study, we looked at DMF’s antitumor effects on ATL cells. Materials and Methods: We examined the effects of DMF on proliferation and apoptosis using the trypan blue exclusion assay and annexin V/propidium iodide staining in HTLV-1-infected and transformed T-cell lines, MT-1 and MT-2 cells. We also evaluated the effects of DMF on the nuclear factor-kappa B (NF-κB) and signal transducers and activators of transcription 3 (STAT3) signaling pathways and anti-apoptotic proteins by immunoblotting. Results: DMF inhibited proliferation and induced apoptosis in MT-1 and MT-2 cells by activating poly ADP-ribose polymerase (PARP). Furthermore, DMF inhibited the constitutive activation of both canonical and non-canonical NF-κB pathways in MT-2 cells and the non-canonical NF-κB pathway in MT-1 cells. DMF also inhibited the constitutive tyrosine phosphorylation of STAT3 and the expression of anti-apoptotic proteins, c-IAP2 and survivin in both cells. Conclusion: These results indicate that DMF inhibits proliferation and induces apoptosis in HTLV-1-infected and transformed T-cells by suppressing NF-κB and STAT3 signaling pathways. DMF should be investigated further as a novel agent for ATL.

Key Words:
  • Dimethyl fumarate
  • NF-κB
  • STAT3
  • c-IAP2
  • adult T-cell
  • leukemia
  • lymphoma

Adult T-cell leukemia (ATL) is a peripheral T lymphocytic malignancy, which is caused by the human T-cell leukemia virus type 1 (HTLV-1) (1-3). HTLV-1 is prevalent worldwide, with the highest known prevalence in Japan, Caribbean islands, South America, the African continent and Melanesia (4). The median age of diagnosis in Japan was 69 years, according to a Japanese nationwide epidemiological study of ATL diagnosed between 2012 and 2013 (5). Due to its resistance to conventional chemotherapy, aggressive ATL, including acute and lymphoma types and chronic types with poor prognostic factors, has a poor prognosis with a median survival duration of 13 months (6). Recently, newer agents have been developed. Mogamulizumab is a humanized anti-CC chemokine receptor type 4 (CCR4) antibody that has been approved in Japan for the treatment of newly diagnosed aggressive ATL in combination with a conventional dose-intensified multidrug regimen, namely, the modified LSG15 (mLSG15) regimen (VCAP-AMP-VECP: vincristine, cyclophosphamide, doxorubicin, and prednisolone; doxorubicin, ranimustine, and prednisolone; vindesine, etoposide, carboplatin, and prednisolone). In a randomized phase II study, the complete response rate as the primary endpoint was significantly higher in the mogamulizumab plus mLSG15 arm (52%) than in LSG15 alone (33%) (7). Despite the development of treatment incorporating such novel agents, most ATL patients relapse and require additional treatment. As a result, new therapeutic agents that effectively kill ATL cells need to be developed.

The proliferation of ATL cells is related to the constitutive activation of the nuclear factor-kappa B (NF-κB) pathway and Janus kinase (JAK) and signal transducers and activators of transcription (STAT) proteins (8, 9). Furthermore, NF-κB is induced by a unique HTLV-1 gene, Tax, which has been linked to tumor growth in an in vivo model of ATL (9). Thus, the activation of various intracellular signaling molecules is critical for HTLV-1 oncogenesis. Many researchers have been working on developing drugs that target these signaling molecules. Dehydroxymethylepoxyquinomicin (DHMEQ), a novel NF-κB inhibitor, inhibited NF-κB activation and induced apoptosis in primary ATL cells and cell lines derived from them (10). A proteasome inhibitor, bortezomib, also suppressed NF-κB activation via inhibiting the degradation of IκBα in ATL cells and suppressed tumor growth in vivo (11). Based on these results, a phase II study of bortezomib in relapsed or refractory aggressive ATL was conducted. Unfortunately, the study was terminated early because the single-agent activity did not appear to be very promising (12). We previously demonstrated that the naturally occurring rotenoid deguelin and the well-known polyphenol compound resveratrol suppressed cell growth and survival via inhibiting STAT3 activation in HTLV-1-infected and transformed cells (13, 14).

Dimethyl fumarate (DMF) is a drug approved by the US Food and Drug Administration and the European Medicines Agency and it is clinically used as first-line treatment of relapsing-remitting multiple sclerosis or as a systemic medication for moderate to severe psoriasis. It has been shown that DMF induces apoptosis in activated T cells (15). DMF is also known as an activator of nuclear factor-erythroid 2-related factor 2 (Nrf2) (16). DMF has shown to enhance the differentiation induced by vitamin D derivatives in acute myeloid leukemia cells and inhibit tumor growth via the vitamin D receptor and Nrf2 signaling (17). It inhibits the NF-κB pathway in breast cancer, cutaneous T-cell lymphoma (CTCL), and diffuse large B-cell lymphoma (DLBCL) cells (18-20). Furthermore, DMF has been shown to inhibit the STAT3 pathway in DLBCL and hepatocellular carcinoma cells, and the Wnt signaling pathway in chronic lymphocytic leukemia cells (18, 21, 22). However, DMF’s antitumor effect and mechanism of action in ATL cells are unknown. In this study, we report that DMF induces cell death in HTLV-1-infected and transformed T cells by suppressing the NF-κB and STAT3 pathways.

Materials and Methods

Reagents and antibodies. Sigma-Aldrich supplied the DMF (Tokyo, Japan). Working solutions were prepared in RPMI 1640 medium from a stock solution prepared by dissolving in dimethyl sulfoxide (DMSO). Control cells were incubated with DMSO (maximum concentration) alone. Antibodies against phospho-STAT3 (Tyr705) (#9145), STAT3 (#4904), phosho-IκBα (Ser32) (#2859), IκBα (#9242), a cellular inhibitor of apoptosis protein-2 (c-IAP2) (#3130), cleaved poly ADP-ribose polymerase (PARP) (#9541), survivin (#28089), Bcl-extra-large (Bcl-xL) (#2764), NF-κB p65 (#6956), and β-actin (#4967) were obtained from Cell Signaling Technology (Beverly, MA). Antibodies against RelB (sc-48366), B-cell lymphoma-2 (Bcl-2) (sc-7382), histone deacetylase 1 (HDAC1) (sc-7872), and NF-κB p52 (sc-7386), and horseradish peroxidase (HRP) -conjugated secondary antibodies (sc-7074, sc-707692) were obtained from Santa Cruz Biotechnology (Dallas, TX). Antibodies against NF-κB p50 (616702) were obtained from Biolegend (San Diego, CA). Primary and secondary antibodies were diluted at 1:1000 and 1:5000, respectively.

Cell lines and culture. The Japanese Collection of Research Bioresources (JCRB) Cell Bank (Tokyo, Japan) provided the MT-1 (a leukemic T-cell line derived from ATL patients) and the MT-2 (a human HTLV-1-infected cell line) cell lines. Both cell lines were cultured in RPMI1640 supplemented with 10% fetal bovine serum (FBS) (Hyclone, Logan, UT), 50 U/mL penicillin (Thermo Fisher Scientific, Tokyo, Japan), and 50 μg/mL streptomycin (Thermo Fisher Scientific) in a 5% CO2 incubator at 37°C.

Cell proliferation and cell death assay. Cell viability was determined using the trypan blue exclusion assay with a Countess II FL Automated Cell Counter after cells were cultured in RPMI1640 medium supplemented with 10% FBS in the presence or absence of DMF for 24 or 48 h, according to the manufacturer’s instructions (Thermo Fisher Scientific, Waltham, MA, USA). The proliferation rate was defined as the percentage of surviving cells in each group compared to that in the untreated group. For apoptosis assay, cells (1×106/ml) were treated in the presence or absence of DMF for 24 or 48 h. Then, cells were stained with annexin V and propidium iodide (PI) according to the manufacturer’s instructions (BD Biosciences, San Diego, CA, USA). The percentage of annexin V-positive cells was analyzed with a flow cytometer (FACSCalibur, BD Biosciences).

Immunoblotting. After being treated with RPMI 1640 supplemented with 10% FBS in the presence or absence of DMF, the MT-1 and MT-2 cells were washed twice with cold PBS. Then, we prepared cell lysates using ice-cold RIPA lysis buffer (Santa Cruz Biotechnology, Dallas, TX, USA) with 5 mM NaF, 0.5 mM sodium orthovanadate, and 1% proteasome inhibitors, incubated for 30 min at 4°C, and then centrifuged at 10,000 rpm for 10 min, while supernatants were retained. The cell lysates were eluted by incubating them for 5 min at 95°C in sodium dodecyl sulfate sample buffer (Bio-Rad Laboratories, Hercules, CA, USa). The samples were loaded into 7-15% Tris-glycine gel (Bio-Rad Laboratories) and transferred to polyvinylidene difluoride membranes (GE Healthcare, Buckinghamshire, UK). Nuclear and cytoplasmic fractions from each extract were prepared using the Cytoplasmic and Nuclear Protein Extraction Kit (101Bio, Palo Alto, CA, USA) according to the manufacturer’s instructions. Next, the immunoblotting assays were performed according to the instructions provided by the manufacturers of the antibodies. Blots were visualized by using horseradish peroxidase-conjugated secondary antibodies and enhanced chemiluminescence detection reagent (GE Healthcare, Tokyo, Japan). Membranes were incubated in stripping buffer (Pierce, Rockford, IL, USA) according to the manufacturer’s instructions to re-blotting with other antibodies.

Statistical analysis. JMP11 was used for the statistical analysis (SAS Institute, Cary, NC, USA). The data obtained were expressed as mean±standard deviation (SD). Statistical differences between test groups were analyzed using ANOVA with Bonferroni’s multiple comparison test after verifying that the data met the assumptions of the statistical test employed. p-Values <0.05 were considered statistically significant.

Results

DMF suppresses cell proliferation and survival in MT-1 and MT-2 cells. We initially examined the effects of DMF treatment and discovered that it significantly inhibited the proliferation of MT-1 and MT-2 cells after 24 and 48 h in a dose-dependent manner as shown in Figure 1A and B. Next, we examined whether DMF induces cell death. However, the sensitivity of these cell lines varied, with MT-2 cells being more sensitive to DMF. Flow cytometric analysis with annexin V and PI staining of cells treated with 50 μM and 100 μM DMF for 24 and 48 h was used to determine the frequency of cell death. The frequency of apoptotic (annexin V-positive) cells was significantly higher in the presence of DMF than in its absence (Figure 2A-D). MT-2 cells were also more sensitive to DMF than MT-1 cells on induction of apoptosis. These results suggest that DMF suppressed cell proliferation and induced apoptosis in both MT-1 and MT-2 cells.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Inhibitory effects of DMF on proliferation in MT-1 and MT-2 cells. (A) MT-1 and (B) MT-2 cells were cultured in 24-well tissue culture plates treated with DMSO as a control and DMF at the indicated concentrations for 24 and 48 h. Cell viability was measured using the trypan blue exclusion assay. Data are expressed as the mean percentage of the control cells representing the average of triplicate samples. Error bars represent one standard deviation (SD) from the mean of the triplicate wells. The results are representative of three independent experiments (DMF vs. control, *p-Value <0.05 using ANOVA with Bonferroni multiple comparison test).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

DMF-mediated apoptosis in MT-1 and MT-2 cells. MT-1 and MT-2 cells were cultured in 24-well tissue culture plates treated with DMSO as a control and DMF at the indicated concentrations for 24 and 48 h. The degree of apoptosis was measured using flow cytometric analysis with annexin V and PI staining. The percentages of annexin V-positive cells are indicative of apoptotic cells. Representative dot plots of (A) MT-1 and (B) MT-2 cells are shown. Numbers in dot plots correspond to percentage of cells. Data representing the average of triplicate samples of (C) MT-1 and (D) MT-2 cells are shown. Error bars represent 1 SD from the mean for triplicate wells. Results are representative of two independent experiments (DMF vs. control, *p-Value <0.05 using ANOVA with Bonferroni multiple comparison test).

DMF inhibits phosphorylation of IκBα and induces PARP activation in MT-1 and MT-2 cells. To better understand the mechanism by which DMF suppresses the proliferation and induces apoptosis of MT-1 and MT-2 cells, we investigated whether DMF inhibits IkBα phosphorylation and induces PARP activation in both cells. IκBα normally binds to the complex of NF-κB p65 and p50, both of which are transcriptional regulators and are responsible for the suppression of nuclear translocation. As shown in Figure 3A, DMF treatment for 24 h inhibited IκBα phosphorylation in MT-1 and MT-2 cells. Furthermore, DMF induced PARP activation in a dose-dependent manner in both cells.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

DMF suppresses the NF-κB signaling pathways and activates PARP in MT-1 and MT-2 cells. (A) MT-1 and MT-2 cells were treated with DMSO and DMF at the indicated concentrations for 24 h. Immunoblotting was performed with specific antibodies against phospho-IκBα at Ser32, IκBα, cleaved PARP and β-actin. (B) MT-1 and MT-2 cells were treated with or without DMF at the indicated concentrations for 48 and 24 h, respectively. Nuclear and cytoplasmic fractions from each extract were prepared as described in Materials and methods. Immunoblotting was performed with specific antibodies against p52, RelB, p65, p50, HDAC1, and β-actin. All the immunoblots shown are representative of two independent experiments. FL-PARP, full-length PARP; CL-PARP, cleaved PARP; D, DMSO.

DMF inhibits nuclear translocation of NF-κB molecules in MT-1 and MT-2 cells. Next, we investigated if DMF suppresses the canonical and non-canonical NF-κB pathway in MT-1 and MT-2 cells. Since 100 μM DMF for 48 h caused a significant increase in apoptosis in MT-1 cells (Figure 2C), MT-1 cells were treated with 50 μM and 100 μM DMF for 48 h. As shown in Figure 3B, treatment with 100 μM DMF suppressed RelB expression in the nucleus of MT-1 cells. On the other hand, MT-2 cells were treated with 50 μM and 100 μM DMF for 24 h. Treatment with at least 50 μM DMF suppressed p52, RelB, and p65 expression in the nucleus of MT-2 cells.

DMF inhibits STAT3 phosphorylation, cIAP-2, and survivin expression in MT-1 and MT-2 cells. STAT3 was found to be phosphorylated all the time in MT-1 and MT-2 cells. DMF treatment for 24 h suppressed STAT3 phosphorylation in MT-1 and MT-2 cells, as shown in Figure 4. We then looked at how DMF affected anti-apoptotic proteins, including c-IAP2, survivin, Bcl-2, and Bcl-xL in both cells. DMF inhibited cIAP-2 and survivin, both of which are known as downstream targets of NF-κB and STAT3, but not Bcl-2 and Bcl-xL in MT-1 and MT-2 cells (Figure 4).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

DMF suppresses the constitutive phosphorylation of STAT3 at Tyr705 and c-IAP2 and survivin protein expression in MT-1 and MT-2 cells. MT-1 and MT-2 cells were treated with DMSO and DMF at the indicated concentrations for 24 h. Immunoblotting was performed with specific antibodies against phospho-STAT3 at Tyr705, STAT3, c-IAP2, survivin, Bcl-xL, Bcl-2 and β-actin. All the immunoblots shown are representative of two independent experiments.

Discussion

In this study, we demonstrated for the first time that DMF inhibited proliferation and survival of HTLV-1-infected and transformed cells. DMF is approved for the treatment of multiple sclerosis and psoriasis. It has been used in several clinical trials for its anti-inflammatory and anti-cancer properties (23). It has been shown that DMF induces cell death in various malignant cells including DLBCL, CTCL, breast cancer, hepatocellular carcinoma, melanoma, glioblastoma, lung cancer, and colorectal cancer (18-21, 24-27). Thus, DMF is a promising drug candidate for cancer therapy. In this study, we discovered that DMF significantly inhibits the proliferation and survival of MT-1 and MT-2 cells after 24 and 48 h (Figure 1 and Figure 2). MT-2 cells were more sensitive to DMF on apoptosis than MT-1 cells, similar to the antiproliferative effect of DMF on both cell lines. These findings could be attributed to differences in cell line characteristics; MT-1 cells are derived from ATL while MT-2 cells are derived from human cord leukocytes co-culturing with ATL cells (28, 29).

HTLV-1 Tax, the viral transcription activator protein, has been shown to activate various signaling pathways via direct interactions with signaling molecules (30). Despite the absence of Tax in primary ATL cells, these signaling pathways are activated, as well as HTLV-1-infected cells, in which Tax is present. These findings suggest that Tax-mediated signaling pathway activation is involved in the early stages of transformation. The NF-κB signaling pathway, among others, is constitutively activated in ATL cells (31). The canonical NF-κB pathway consists of p65 and p50 transcription factors, both of which are held in the cytoplasm by an inhibitor protein, IκBα. Phosphorylation of IκBα by upstream kinases leads to proteasomal degradation of IκBα. As a result, p65/p50 molecules can translocate to the nucleus, where they bind to DNA and induce gene transcription (32). The non-canonical NF-κB pathway consists of RelB and p52 transcription factors. This pathway is dependent on phosphorylation-induced p100 processing, which is activated by signaling from a subset of tumor necrosis factor receptors. The processing of p100 results in the production of p52 and the nuclear translocation of the RelB/p52 heterodimer (33, 34).

In this regard, DMF was tested to see if it inhibited the activation of the NF-κB pathway in MT-1 and MT-2 cells. DMF inhibited constitutive phosphorylation of IκB in MT-1 and MT-2 cells, as shown in Figure 3A. p65, p50, RelB, and p52 molecules were found in the nuclear fraction of MT-2 cells, indicating that both canonical and non-canonical pathways are active in MT-2 cells (Figure 3B). In addition, DMF suppressed p65, RelB, and p52 expression in the nucleus in a dose-dependent manner in MT-2 cells (Figure 3B). These results suggest that DMF inhibits both canonical and non-canonical NF-κB pathways in MT-2 cells. On the other hand, in MT-1 cells, we did not detect p65 molecule in the nucleus before treatment with DMF, suggesting that the canonical pathway is not activated in MT-1 cells (Figure 3B). Only RelB expression in the nucleus was suppressed by treatment with 100 μM DMF for 48 h in MT-1 cells (Figure 3B). DMF inhibits the non-canonical NF-κB pathway in MT-1 cells, according to these findings. However, DMF’s inhibitory effect on NF-κB pathway molecules was significantly weaker in MT-1 cells than in MT-2 cells. The difference appeared to parallel the effect of DMF on cell proliferation and apoptosis levels in MT-1 and MT-2 cells. DMF has recently been shown to induce cell death by inhibiting the NF-κB pathway in CTCL and DLBCL (18, 20). DMF induced cell death in CTCL cells and inhibited CTCL tumor growth and metastasis in vivo, according to Nicolay et al. (20). They also demonstrated that DNA binding of p50, p65, and RelB proteins was inhibited after DMF treatment in CTCL cell lines. Schmitt et al. showed that DMF down-regulated several NF-κB gene signatures using gene set enrichment analysis in activated B-cell-like (ABC) DLBCL (18). Furthermore, they demonstrated that DMF treatment impaired the phosphorylation of IκBα at S32/36, indicating that DMF disrupted the chronic activation of the IκB kinase (IKK) complex. As expected, DMF treatment resulted in markedly decreased p65 translocation to the nucleus and in impaired binding of p65 and c-Rel to their consensus DNA sequence (18). These findings are consistent with our findings in MT-2 cells, which show that DMF strongly inhibits the NF-κB pathway in HTLV-1-infected T cells, MT-2. More research is needed to determine the precise mechanism by which DMF inhibits IκB phosphorylation in ATL cells.

Activation of the JAK/STAT signaling pathway is associated with cell proliferation in ATL (8). We found that DMF treatment simultaneously decreased the constitutive STAT3 phosphorylation in MT-1 and MT-2 cells (Figure 4), indicating a direct role of DMF in STAT3 inhibition. DMF inhibited STAT3 phosphorylation in ABC DLBCL, according to Schmitt et al. (18). DMF’s direct modification and inhibition of JAK1 and TYK2 were also identified as another mechanism for its toxicity in ABC DLBCL. Future research should focus on determining the precise mechanism by which DMF inhibits STAT3 phosphorylation in ATL cells. We also discovered that DMF inhibited c-IAP2 and survivin protein expression in both cell lines (Figure 4). Both antiapoptotic proteins have been previously reported as an NF-κB target and STAT3 (13, 14, 35). A recent study showed that NF-κB and IRF4 regulated several cancer genes such as MYC, CCR4, and c-IAP2 in ATL cells (36). These findings indicate that DMF induces apoptosis by inhibiting c-IAP2 and survivin expression.

Overall, by suppressing the NF-κB and STAT3 signaling pathways, DMF inhibits proliferation and induces apoptosis in HTLV-1-infected and transformed T cells. DMF should be investigated further as a novel ATL agent.

Acknowledgements

We thank Ms. Yuki Seki for her technical assistance. We appreciate Enago for English language editing. This study was supported by a research grant (20113 [S.I.] from the Japanese Society of Hematology).

Footnotes

  • Authors’ Contributions

    All Authors performed experiments. T.M. and S.I. designed the research, wrote the paper and discussed the manuscript.

  • Conflicts of Interest

    The Authors declare no competing financial interests.

  • Received February 10, 2022.
  • Revision received April 9, 2022.
  • Accepted April 11, 2022.
  • Copyright © 2022 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).

References

  1. ↵
    1. Poiesz BJ,
    2. Ruscetti FW,
    3. Gazdar AF,
    4. Bunn PA,
    5. Minna JD and
    6. Gallo RC
    : Detection and isolation of type C retrovirus particles from fresh and cultured lymphocytes of a patient with cutaneous T-cell lymphoma. Proc Natl Acad Sci USA 77(12): 7415-7419, 1980. PMID: 6261256. DOI: 10.1073/pnas.77.12.7415
    OpenUrlAbstract/FREE Full Text
    1. Hinuma Y,
    2. Nagata K,
    3. Hanaoka M,
    4. Nakai M,
    5. Matsumoto T,
    6. Kinoshita KI,
    7. Shirakawa S and
    8. Miyoshi I
    : Adult T-cell leukemia: antigen in an ATL cell line and detection of antibodies to the antigen in human sera. Proc Natl Acad Sci USA 78(10): 6476-6480, 1981. PMID: 7031654. DOI: 10.1073/pnas.78.10.6476
    OpenUrlAbstract/FREE Full Text
  2. ↵
    1. Yoshida M,
    2. Miyoshi I and
    3. Hinuma Y
    : Isolation and characterization of retrovirus from cell lines of human adult T-cell leukemia and its implication in the disease. Proc Natl Acad Sci USA 79(6): 2031-2035, 1982. PMID: 6979048. DOI: 10.1073/pnas.79.6.2031
    OpenUrlAbstract/FREE Full Text
  3. ↵
    1. Gessain A and
    2. Cassar O
    : Epidemiological aspects and world distribution of HTLV-1 infection. Front Microbiol 3: 388, 2012. PMID: 23162541. DOI: 10.3389/fmicb.2012.00388
    OpenUrlCrossRefPubMed
  4. ↵
    1. Ito S,
    2. Iwanaga M,
    3. Nosaka K,
    4. Imaizumi Y,
    5. Ishitsuka K,
    6. Amano M,
    7. Utsunomiya A,
    8. Tokura Y,
    9. Watanabe T,
    10. Uchimaru K,
    11. Tsukasaki K and Collaborative Investigators
    : Epidemiology of adult T-cell leukemia-lymphoma in Japan: An updated analysis, 2012-2013. Cancer Sci 112(10): 4346-4354, 2021. PMID: 34355480. DOI: 10.1111/cas.15097
    OpenUrlCrossRefPubMed
  5. ↵
    1. Yamada Y,
    2. Tomonaga M,
    3. Fukuda H,
    4. Hanada S,
    5. Utsunomiya A,
    6. Tara M,
    7. Sano M,
    8. Ikeda S,
    9. Takatsuki K,
    10. Kozuru M,
    11. Araki K,
    12. Kawano F,
    13. Niimi M,
    14. Tobinai K,
    15. Hotta T and
    16. Shimoyama M
    : A new G-CSF-supported combination chemotherapy, LSG15, for adult T-cell leukaemia-lymphoma: Japan Clinical Oncology Group Study 9303. Br J Haematol 113(2): 375-382, 2001. PMID: 11380402. DOI: 10.1046/j.1365-2141.2001.02737.x
    OpenUrlCrossRefPubMed
  6. ↵
    1. Ishida T,
    2. Jo T,
    3. Takemoto S,
    4. Suzushima H,
    5. Uozumi K,
    6. Yamamoto K,
    7. Uike N,
    8. Saburi Y,
    9. Nosaka K,
    10. Utsunomiya A,
    11. Tobinai K,
    12. Fujiwara H,
    13. Ishitsuka K,
    14. Yoshida S,
    15. Taira N,
    16. Moriuchi Y,
    17. Imada K,
    18. Miyamoto T,
    19. Akinaga S,
    20. Tomonaga M and
    21. Ueda R
    : Dose-intensified chemotherapy alone or in combination with mogamulizumab in newly diagnosed aggressive adult T-cell leukaemia-lymphoma: a randomized phase II study. Br J Haematol 169(5): 672-682, 2015. PMID: 25733162. DOI: 10.1111/bjh.13338
    OpenUrlCrossRefPubMed
  7. ↵
    1. Takemoto S,
    2. Mulloy JC,
    3. Cereseto A,
    4. Migone TS,
    5. Patel BK,
    6. Matsuoka M,
    7. Yamaguchi K,
    8. Takatsuki K,
    9. Kamihira S,
    10. White JD,
    11. Leonard WJ,
    12. Waldmann T and
    13. Franchini G
    : Proliferation of adult T cell leukemia/lymphoma cells is associated with the constitutive activation of JAK/STAT proteins. Proc Natl Acad Sci USA 94(25): 13897-13902, 1997. PMID: 9391124. DOI: 10.1073/pnas.94.25.13897
    OpenUrlAbstract/FREE Full Text
  8. ↵
    1. Dewan MZ,
    2. Terashima K,
    3. Taruishi M,
    4. Hasegawa H,
    5. Ito M,
    6. Tanaka Y,
    7. Mori N,
    8. Sata T,
    9. Koyanagi Y,
    10. Maeda M,
    11. Kubuki Y,
    12. Okayama A,
    13. Fujii M and
    14. Yamamoto N
    : Rapid tumor formation of human T-cell leukemia virus type 1-infected cell lines in novel NOD-SCID/gammac(null) mice: suppression by an inhibitor against NF-kappaB. J Virol 77(9): 5286-5294, 2003. PMID: 12692230. DOI: 10.1128/jvi.77.9.5286-5294.2003
    OpenUrlAbstract/FREE Full Text
  9. ↵
    1. Watanabe M,
    2. Ohsugi T,
    3. Shoda M,
    4. Ishida T,
    5. Aizawa S,
    6. Maruyama-Nagai M,
    7. Utsunomiya A,
    8. Koga S,
    9. Yamada Y,
    10. Kamihira S,
    11. Okayama A,
    12. Kikuchi H,
    13. Uozumi K,
    14. Yamaguchi K,
    15. Higashihara M,
    16. Umezawa K,
    17. Watanabe T and
    18. Horie R
    : Dual targeting of transformed and untransformed HTLV-1-infected T cells by DHMEQ, a potent and selective inhibitor of NF-kappaB, as a strategy for chemoprevention and therapy of adult T-cell leukemia. Blood 106(7): 2462-2471, 2005. PMID: 15956280. DOI: 10.1182/blood-2004-09-3646
    OpenUrlAbstract/FREE Full Text
  10. ↵
    1. Satou Y,
    2. Nosaka K,
    3. Koya Y,
    4. Yasunaga JI,
    5. Toyokuni S and
    6. Matsuoka M
    : Proteasome inhibitor, bortezomib, potently inhibits the growth of adult T-cell leukemia cells both in vivo and in vitro. Leukemia 18(8): 1357-1363, 2004. PMID: 15190257. DOI: 10.1038/sj.leu.2403400
    OpenUrlCrossRefPubMed
  11. ↵
    1. Ishitsuka K,
    2. Utsunomiya A,
    3. Katsuya H,
    4. Takeuchi S,
    5. Takatsuka Y,
    6. Hidaka M,
    7. Sakai T,
    8. Yoshimitsu M,
    9. Ishida T and
    10. Tamura K
    : A phase II study of bortezomib in patients with relapsed or refractory aggressive adult T-cell leukemia/lymphoma. Cancer Sci 106(9): 1219-1223, 2015. PMID: 26179770. DOI: 10.1111/cas.12735
    OpenUrlCrossRefPubMed
  12. ↵
    1. Ito S,
    2. Oyake T,
    3. Murai K and
    4. Ishida Y
    : Deguelin suppresses cell proliferation via the inhibition of survivin expression and STAT3 phosphorylation in HTLV-1-transformed T cells. Leuk Res 34(3): 352-357, 2010. PMID: 19781773. DOI: 10.1016/j.leukres.2009.09.003
    OpenUrlCrossRefPubMed
  13. ↵
    1. Suzuki Y,
    2. Ito S,
    3. Sasaki R,
    4. Asahi M and
    5. Ishida Y
    : Resveratrol suppresses cell proliferation via inhibition of STAT3 phosphorylation and Mcl-1 and cIAP-2 expression in HTLV-1-infected T cells. Leuk Res 37(12): 1674-1679, 2013. PMID: 24090995. DOI: 10.1016/j.leukres.2013.09.010
    OpenUrlCrossRefPubMed
  14. ↵
    1. Treumer F,
    2. Zhu K,
    3. Gläser R and
    4. Mrowietz U
    : Dimethylfumarate is a potent inducer of apoptosis in human T cells. J Invest Dermatol 121(6): 1383-1388, 2003. PMID: 14675187. DOI: 10.1111/j.1523-1747.2003.12605.x
    OpenUrlCrossRefPubMed
  15. ↵
    1. Cuadrado A,
    2. Manda G,
    3. Hassan A,
    4. Alcaraz MJ,
    5. Barbas C,
    6. Daiber A,
    7. Ghezzi P,
    8. León R,
    9. López MG,
    10. Oliva B,
    11. Pajares M,
    12. Rojo AI,
    13. Robledinos-Antón N,
    14. Valverde AM,
    15. Guney E and
    16. Schmidt HHHW
    : Transcription factor NRF2 as a therapeutic target for chronic diseases: a systems medicine approach. Pharmacol Rev 70(2): 348-383, 2018. PMID: 29507103. DOI: 10.1124/pr.117.014753
    OpenUrlAbstract/FREE Full Text
  16. ↵
    1. Nachliely M,
    2. Trachtenberg A,
    3. Khalfin B,
    4. Nalbandyan K,
    5. Cohen-Lahav M,
    6. Yasuda K,
    7. Sakaki T,
    8. Kutner A and
    9. Danilenko M
    : Dimethyl fumarate and vitamin D derivatives cooperatively enhance VDR and Nrf2 signaling in differentiating AML cells in vitro and inhibit leukemia progression in a xenograft mouse model. J Steroid Biochem Mol Biol 188: 8-16, 2019. PMID: 30508646. DOI: 10.1016/j.jsbmb.2018.11.017
    OpenUrlCrossRefPubMed
  17. ↵
    1. Schmitt A,
    2. Xu W,
    3. Bucher P,
    4. Grimm M,
    5. Konantz M,
    6. Horn H,
    7. Zapukhlyak M,
    8. Berning P,
    9. Brändle M,
    10. Jarboui MA,
    11. Schönfeld C,
    12. Boldt K,
    13. Rosenwald A,
    14. Ott G,
    15. Grau M,
    16. Klener P,
    17. Vockova P,
    18. Lengerke C,
    19. Lenz G,
    20. Schulze-Osthoff K and
    21. Hailfinger S
    : Dimethyl fumarate induces ferroptosis and impairs NF-κB/STAT3 signaling in DLBCL. Blood 138(10): 871-884, 2021. PMID: 33876201. DOI: 10.1182/blood.2020009404
    OpenUrlCrossRefPubMed
    1. Kastrati I,
    2. Siklos MI,
    3. Calderon-Gierszal EL,
    4. El-Shennawy L,
    5. Georgieva G,
    6. Thayer EN,
    7. Thatcher GR and
    8. Frasor J
    : Dimethyl fumarate inhibits the nuclear factor κB pathway in breast cancer cells by covalent modification of p65 protein. J Biol Chem 291(7): 3639-3647, 2016. PMID: 26683377. DOI: 10.1074/jbc.M115.679704
    OpenUrlAbstract/FREE Full Text
  18. ↵
    1. Nicolay JP,
    2. Müller-Decker K,
    3. Schroeder A,
    4. Brechmann M,
    5. Möbs M,
    6. Géraud C,
    7. Assaf C,
    8. Goerdt S,
    9. Krammer PH and
    10. Gülow K
    : Dimethyl fumarate restores apoptosis sensitivity and inhibits tumor growth and metastasis in CTCL by targeting NF-κB. Blood 128(6): 805-815, 2016. PMID: 27268084. DOI: 10.1182/blood-2016-01-694117
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Liu H,
    2. Feng XD,
    3. Yang B,
    4. Tong RL,
    5. Lu YJ,
    6. Chen DY,
    7. Zhou L,
    8. Xie HY,
    9. Zheng SS and
    10. Wu J
    : Dimethyl fumarate suppresses hepatocellular carcinoma progression via activating SOCS3/JAK1/STAT3 signaling pathway. Am J Transl Res 11(8): 4713-4725, 2019. PMID: 31497193.
    OpenUrlPubMed
  20. ↵
    1. Wu C,
    2. Lao F,
    3. Li H,
    4. Rassenti L,
    5. Kipps T,
    6. Messer K,
    7. Carson D and
    8. Choi M
    : Inhibition of Wnt signaling by dimethyl fumarate results in in vitro and in vivo clearance of chronic lymphocytic leukemia cells and has additive activity with ibrutinib. Blood 124(21): 4683-4683, 2019. DOI: 10.1182/blood.V124.21.4683.4683
    OpenUrlCrossRef
  21. ↵
    1. Saidu NEB,
    2. Kavian N,
    3. Leroy K,
    4. Jacob C,
    5. Nicco C,
    6. Batteux F and
    7. Alexandre J
    : Dimethyl fumarate, a two-edged drug: Current status and future directions. Med Res Rev 39(5): 1923-1952, 2019. PMID: 30756407. DOI: 10.1002/med.21567
    OpenUrlCrossRefPubMed
  22. ↵
    1. Loewe R,
    2. Valero T,
    3. Kremling S,
    4. Pratscher B,
    5. Kunstfeld R,
    6. Pehamberger H and
    7. Petzelbauer P
    : Dimethylfumarate impairs melanoma growth and metastasis. Cancer Res 66(24): 11888-11896, 2006. PMID: 17178886. DOI: 10.1158/0008-5472.CAN-06-2397
    OpenUrlAbstract/FREE Full Text
    1. Ghods AJ,
    2. Glick R,
    3. Braun D and
    4. Feinstein D
    : Beneficial actions of the anti-inflammatory dimethyl fumarate in glioblastomas. Surg Neurol Int 4: 160, 2013. PMID: 24404403. DOI: 10.4103/2152-7806.123656
    OpenUrlCrossRefPubMed
    1. Saidu NE,
    2. Noé G,
    3. Cerles O,
    4. Cabel L,
    5. Kavian-Tessler N,
    6. Chouzenoux S,
    7. Bahuaud M,
    8. Chéreau C,
    9. Nicco C,
    10. Leroy K,
    11. Borghese B,
    12. Goldwasser F,
    13. Batteux F and
    14. Alexandre J
    : Dimethyl fumarate controls the NRF2/DJ-1 axis in cancer cells: therapeutic applications. Mol Cancer Ther 16(3): 529-539, 2017. PMID: 28069874. DOI: 10.1158/1535-7163.MCT-16-0405
    OpenUrlAbstract/FREE Full Text
  23. ↵
    1. Xie X,
    2. Zhao Y,
    3. Ma CY,
    4. Xu XM,
    5. Zhang YQ,
    6. Wang CG,
    7. Jin J,
    8. Shen X,
    9. Gao JL,
    10. Li N,
    11. Sun ZJ and
    12. Dong DL
    : Dimethyl fumarate induces necroptosis in colon cancer cells through GSH depletion/ROS increase/MAPKs activation pathway. Br J Pharmacol 172(15): 3929-3943, 2015. PMID: 25953698. DOI: 10.1111/bph.13184
    OpenUrlCrossRefPubMed
  24. ↵
    1. Miyoshi I,
    2. Kubonishi I,
    3. Sumida M,
    4. Hiraki S,
    5. Tsubota T,
    6. Kimura I,
    7. Miyamoto K and
    8. Sato J
    : A novel T-cell line derived from adult T-cell leukemia. Gan 71(1): 155-156, 1980. PMID: 6966589.
    OpenUrlPubMed
  25. ↵
    1. Miyoshi I,
    2. Kubonishi I,
    3. Yoshimoto S,
    4. Akagi T,
    5. Ohtsuki Y,
    6. Shiraishi Y,
    7. Nagata K and
    8. Hinuma Y
    : Type C virus particles in a cord T-cell line derived by co-cultivating normal human cord leukocytes and human leukaemic T cells. Nature 294(5843): 770-771, 1981. PMID: 6275274. DOI: 10.1038/294770a0
    OpenUrlCrossRefPubMed
  26. ↵
    1. Yamagishi M and
    2. Watanabe T
    : Molecular hallmarks of adult T cell leukemia. Front Microbiol 3: 334, 2012. PMID: 23060864. DOI: 10.3389/fmicb.2012.00334
    OpenUrlCrossRefPubMed
  27. ↵
    1. Mori N,
    2. Fujii M,
    3. Ikeda S,
    4. Yamada Y,
    5. Tomonaga M,
    6. Ballard DW and
    7. Yamamoto N
    : Constitutive activation of NF-kappaB in primary adult T-cell leukemia cells. Blood 93(7): 2360-2368, 1999. PMID: 10090947.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    1. Hayden MS and
    2. Ghosh S
    : Shared principles in NF-kappaB signaling. Cell 132(3): 344-362, 2008. PMID: 18267068. DOI: 10.1016/j.cell.2008.01.020
    OpenUrlCrossRefPubMed
  29. ↵
    1. Coope HJ,
    2. Atkinson PG,
    3. Huhse B,
    4. Belich M,
    5. Janzen J,
    6. Holman MJ,
    7. Klaus GG,
    8. Johnston LH and
    9. Ley SC
    : CD40 regulates the processing of NF-kappaB2 p100 to p52. EMBO J 21(20): 5375-5385, 2002. PMID: 12374738. DOI: 10.1093/emboj/cdf542
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Derudder E,
    2. Dejardin E,
    3. Pritchard LL,
    4. Green DR,
    5. Korner M and
    6. Baud V
    : RelB/p50 dimers are differentially regulated by tumor necrosis factor-alpha and lymphotoxin-beta receptor activation: critical roles for p100. J Biol Chem 278(26): 23278-23284, 2003. PMID: 12709443. DOI: 10.1074/jbc.M300106200
    OpenUrlAbstract/FREE Full Text
  31. ↵
    1. Wang CY,
    2. Mayo MW,
    3. Korneluk RG,
    4. Goeddel DV and
    5. Baldwin AS Jr.
    : NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science 281(5383): 1680-1683, 1998. PMID: 9733516. DOI: 10.1126/science.281.5383.1680
    OpenUrlAbstract/FREE Full Text
  32. ↵
    1. Wong RWJ,
    2. Tan TK,
    3. Amanda S,
    4. Ngoc PCT,
    5. Leong WZ,
    6. Tan SH,
    7. Asamitsu K,
    8. Hibi Y,
    9. Ueda R,
    10. Okamoto T,
    11. Ishida T,
    12. Iida S and
    13. Sanda T
    : Feed-forward regulatory loop driven by IRF4 and NF-κB in adult T-cell leukemia/lymphoma. Blood 135(12): 934-947, 2020. PMID: 31972002. DOI: 10.1182/blood.2019002639
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 42 (5)
Anticancer Research
Vol. 42, Issue 5
May 2022
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Dimethyl Fumarate Induces Apoptosis via Inhibiting NF-κB and STAT3 Signaling in Adult T-cell Leukemia/Lymphoma Cells
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
1 + 4 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Dimethyl Fumarate Induces Apoptosis via Inhibiting NF-κB and STAT3 Signaling in Adult T-cell Leukemia/Lymphoma Cells
TAKAHIRO MAETA, TSUYOSHI SATO, KAZUYA ASANO, SHIGEKI ITO
Anticancer Research May 2022, 42 (5) 2301-2309; DOI: 10.21873/anticanres.15709

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Dimethyl Fumarate Induces Apoptosis via Inhibiting NF-κB and STAT3 Signaling in Adult T-cell Leukemia/Lymphoma Cells
TAKAHIRO MAETA, TSUYOSHI SATO, KAZUYA ASANO, SHIGEKI ITO
Anticancer Research May 2022, 42 (5) 2301-2309; DOI: 10.21873/anticanres.15709
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • The Prognostic Significance of p16 and its Role as a Surrogate Marker for Human Papilloma Virus in Oral Squamous Cell Carcinoma: An Analysis of 281 Cases
  • Diagnostic Value of Circulating Cell-free DNA in Patients With Papillary Thyroid Cancer
  • Changes in Lysophospholipid Components in Ulcerative Colitis and Colitis-associated Cancer
Show more Experimental Studies

Similar Articles

Keywords

  • Dimethyl fumarate
  • NF-κB
  • STAT3
  • c-IAP2
  • adult T-cell
  • leukemia
  • lymphoma
Anticancer Research

© 2022 Anticancer Research

Powered by HighWire