1
|
Kashyap A, Dai J, Ni X. Therapeutic Targeting of the Janus Kinase/Signal Transducer and Activator of Transcription Pathway in Cutaneous T-Cell Lymphoma. Cancers (Basel) 2025; 17:568. [PMID: 40002165 PMCID: PMC11853177 DOI: 10.3390/cancers17040568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Cutaneous T-cell lymphoma (CTCL) is a rare group of non-Hodgkin lymphomas characterized by the clonal expansion of malignant T cells. While current treatments can alleviate symptoms and significant progress has been made in treating leukemic CTCL, a definitive cure remains elusive. Dysregulation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway is a key driver of CTCL pathogenesis. As a result, therapeutic strategies targeting JAK/STAT signaling have gained momentum, with the increasing use of JAK inhibitors and other agents that effectively suppress this pathway. These immune-modulating therapies have broad effects on physiological processes, inflammation, and the pathological changes associated with both inflammatory diseases and cancers. Several JAK inhibitors, originally FDA-approved for inflammatory conditions, are now being investigated for cancer treatment. Methods: In this paper, a brief review of the literature on JAK/STAT pathway dysregulation in CTCL is provided, highlighting both clinical and preclinical studies involving JAK inhibitors and other agents that target this pathway. Results: Specifically, we focus on six JAK inhibitors currently under clinical investigation-golidocitinib, ruxolitinib, cerdulatinib, tofacitinib, upadacitinib, and abrocitinib. Additionally, we discuss preclinical studies that explore the mechanisms underlying JAK/STAT pathway inhibition in CTCL. Furthermore, we review reported cases in which CTCL relapsed or emerged following JAK inhibitor treatment. Conclusions: Collectively, these findings support the potential clinical utility of targeting the JAK/STAT pathway in CTCL. However, further research is needed to evaluate safety risks, minimize adverse effects, and optimize these therapeutic strategies.
Collapse
Affiliation(s)
- Alisha Kashyap
- John P. and Kathrine G. McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Julia Dai
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiao Ni
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
2
|
Ye MT, Zuo Z, Calin S, Ye F, He H, Kamata W, Yang Y, You MJ. Integrated Clinical Genotype-Phenotype Characteristics of STAT3-Mutated Myeloid Neoplasms. Clin Cancer Res 2024; 30:4681-4689. [PMID: 38767620 DOI: 10.1158/1078-0432.ccr-24-0066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/25/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
PURPOSE STAT3 is a key transcription factor that mediates cancer progression through phosphorylation or gain-of-function mutations. STAT3 activation in myeloid neoplasms (MN) is primarily mediated through phosphorylation. STAT3 mutation has only rarely been reported in MNs. EXPERIMENTAL DESIGN We assessed the clinicopathologic and molecular genetic features of 32 STAT3-mutated MNs. RESULTS The frequency of STAT3 mutation in MNs was <0.5%. Twenty (62.5%) cases were classified as acute myeloid leukemia, 7 (21.9%) as myelodysplastic syndrome, and 5 (15.6%) as chronic myelomonocytic leukemia, but none as myeloproliferative neoplasms. STAT3 mutations occurred at initial diagnosis in 22 (88%) cases or at relapse or upon leukemic transformation. Clonal hierarchy analysis revealed that STAT3 mutations represented the dominant clone in 30% of acute myeloid leukemia cases but were subclonal in myelodysplastic syndrome and chronic myelomonocytic leukemia. Most were missense mutations located at the SH2 domain, Y640F being the most common. STAT3 mutation was accompanied by coexisting mutations in all cases, most frequently SRSF2, TET2, ASXL1, and SETBP1. STAT3 mutations were usually associated with morphologic dysplasia, increased blasts, and monosomy 7/del7q. With a median follow-up of 24.5 months, 21 patients died, 6 had persistent disease, and 5 achieved complete remission after stem cell transplantation. CONCLUSIONS STAT3 mutation is present in various MNs but not in myeloproliferative neoplasms. It is often an early event or occurs upon leukemic transformation, which suggests an important role in the pathogenesis and progression of MNs by activating the JAK-STAT pathway. It may help determine a subset of patients with MNs who may benefit from targeted therapy. See related commentary by Hochman and Frank, p. 4554.
Collapse
MESH Headings
- Humans
- STAT3 Transcription Factor/genetics
- Middle Aged
- Aged
- Mutation
- Male
- Female
- Adult
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/pathology
- Aged, 80 and over
- Genetic Association Studies
- Phenotype
- Myeloproliferative Disorders/genetics
- Myeloproliferative Disorders/pathology
- Myeloproliferative Disorders/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myelomonocytic, Chronic/genetics
- Leukemia, Myelomonocytic, Chronic/pathology
- Genotype
Collapse
Affiliation(s)
- Matthew T Ye
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhuang Zuo
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steliana Calin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fengxi Ye
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hua He
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wataru Kamata
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yaling Yang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - M James You
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
3
|
Wang LL, Wang H, Lin SJ, Xu XY, Hu WJ, Liu J, Zhang HY. ABBV-744 alleviates LPS-induced neuroinflammation via regulation of BATF2-IRF4-STAT1/3/5 axis. Acta Pharmacol Sin 2024; 45:2077-2091. [PMID: 38862817 PMCID: PMC11420366 DOI: 10.1038/s41401-024-01318-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/17/2024] [Indexed: 06/13/2024]
Abstract
Suppression of neuroinflammation using small molecule compounds targeting the key pathways in microglial inflammation has attracted great interest. Recently, increasing attention has been gained to the role of the second bromodomain (BD2) of the bromodomain and extra-terminal (BET) proteins, while its effect and molecular mechanism on microglial inflammation has not yet been explored. In this study, we evaluated the therapeutic effects of ABBV-744, a BD2 high selective BET inhibitor, on lipopolysaccharide (LPS)-induced microglial inflammation in vitro and in vivo, and explored the key pathways by which ABBV-744 regulated microglia-mediated neuroinflammation. We found that pretreatment of ABBV-744 concentration-dependently inhibited the expression of LPS-induced inflammatory mediators/enzymes including NO, TNF-α, IL-1β, IL-6, iNOS, and COX-2 in BV-2 microglial cells. These effects were validated in LPS-treated primary microglial cells. Furthermore, we observed that administration of ABBV-744 significantly alleviated LPS-induced activation of microglia and transcriptional levels of pro-inflammatory factors TNF-α and IL-1β in mouse hippocampus and cortex. RNA-Sequencing (RNA-seq) analysis revealed that ABBV-744 induced 508 differentially expressed genes (DEGs) in LPS-stimulated BV-2 cells, and gene enrichment and gene expression network analysis verified its regulation on activated microglial genes and inflammatory pathways. We demonstrated that pretreatment of ABBV-744 significantly reduced the expression levels of basic leucine zipper ATF-like transcription factor 2 (BATF2) and interferon regulatory factor 4 (IRF4), and suppressed JAK-STAT signaling pathway in LPS-stimulated BV-2 cells and mice, suggesting that the anti-neuroinflammatory effect of ABBV-744 might be associated with regulation of BATF2-IRF4-STAT1/3/5 pathway, which was confirmed by gene knockdown experiments. This study demonstrates the effect of a BD2 high selective BET inhibitor, ABBV-744, against microglial inflammation, and reveals a BATF2-IRF4-STAT1/3/5 pathway in regulation of microglial inflammation, which might provide new clues for discovery of effective therapeutic strategy against neuroinflammation.
Collapse
Affiliation(s)
- Le-le Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Huan Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Si-Jin Lin
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xing-Yu Xu
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wen-Juan Hu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jia Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hai-Yan Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
4
|
Li M, Mo J, Wu D, He H, Hu P. Treadmill training improves neural function recovery in rats with spinal cord injury via JAK2/STAT3 signaling pathway and attenuating apoptosis. Neuroreport 2024; 35:811-821. [PMID: 38973489 DOI: 10.1097/wnr.0000000000002062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
To investigate the role of JAK2/STAT3 signaling pathway in neural function recovery in rats with spinal cord injury (SCI) after treadmill training. Sprague-Dawley rats were randomly divided into four groups: (a) sham group; (b) SCI group; (c) SCI+treadmill training group (SCI/TT); and (d) SCI/TT+AG490 group (a JAK2 inhibitor) ( n = 12). The 12 Sprague-Dawley rats in each group were randomly assigned into 1 st , 3 rd , 7 th , and 14 th day subgroups. The Basso-Beattie-Bresnahan (BBB) locomotor rating scale was used to assess the spinal cord function, and JAK2, STAT3, and IL-6 protein expressions in the rat spinal cord were evaluated by western blot. The level of cell apoptosis and expressions of apoptotic proteins were evaluated by TUNEL assay and immunohistochemistry, respectively. Rats in the SCI+TT group showed a significantly higher BBB score after SCI compared with the SCI group and the SCI/TT+AG490 group. Mechanistically, the JAK2/STAT3 signal pathway was immediately activated after SCI compared with sham group, and JAK2 and STAT3 were obviously upregulated when treadmill training was performed ( P < 0.05). Results of TUNEL assay showed that the apoptotic rate in SCI/TT was significantly lower than that in the SCI group and SCI/TT+AG490 group ( P < 0.05). Besides, the IL-6 expression in the SCI/TT group was significantly attenuated compared with the SCI group ( P < 0.05). Our results showed that physical treadmill training can enhance activation of JAK2/STAT3 signal pathway and attenuate apoptosis in the injured spinal cord, resulting in better functional recovery. These results underline the importance of synergistic treatment strategies for SCI.
Collapse
Affiliation(s)
- Meng Li
- Department of Hyperbaric Oxygen, Zhujiang Hospital, Southern Medical University, Guangzhou
| | - Jinfeng Mo
- Neurology Department, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guangxi
| | - Deguang Wu
- Department of Traumatic Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou
| | - Haibo He
- Department of Traumatic Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou
| | - Panyong Hu
- Department of Spinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guangxi, China
| |
Collapse
|
5
|
Xue Y, Zhang L, Chu L, Song Z, Zhang B, Su X, Liu W, Li X. JAK2/STAT3 Pathway Inhibition by AG490 Ameliorates Experimental Autoimmune Encephalomyelitis via Regulation of Th17 Cells and Autophagy. Neuroscience 2024; 552:65-75. [PMID: 38885894 DOI: 10.1016/j.neuroscience.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024]
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory condition affecting the central nervous system, and experimental autoimmune encephalomyelitis (EAE) animal models have been extensively used to study it. T-helper 17 cells, which produce interleukin-17(IL-17), play crucial roles in MS pathogenesis, and the JAK2/STAT3 pathway has an essential function in their differentiation from naive CD4 + T cells. This study investigated the effects of the JAK2/STAT3 pathway inhibitor AG490 on EAE in vivo and in vitro, as well as the underlying mechanisms. AG490 ameliorated EAE severity and attenuated its typical symptoms by downregulating proteins associated with the JAK2/STAT3 pathway. Furthermore, it decreased T-helper 17 cell differentiation from naive CD4 + T cells by inactivating STAT3. In addition, it conferred protective effects against EAE by restoring autophagy. These findings indicate the potential of AG490 as a candidate anti-MS therapeutic.
Collapse
Affiliation(s)
- Yumei Xue
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Lu Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lifang Chu
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Zhe Song
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Bing Zhang
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Xiaohui Su
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Wanhu Liu
- Department of Neurology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaobing Li
- Department of Pharmacy, Shijiazhuang People's Hospital, Shijiazhuang, China.
| |
Collapse
|
6
|
Shih BB, Ma C, Cortes JR, Reglero C, Miller H, Quinn SA, Albero R, Laurent AP, Mackey A, Ferrando AA, Geskin L, Palomero T. Romidepsin and Afatinib Abrogate Jak-Signal Transducer and Activator of Transcription Signaling and Elicit Synergistic Antitumor Effects in Cutaneous T-Cell Lymphoma. J Invest Dermatol 2024; 144:1579-1589.e8. [PMID: 38219917 PMCID: PMC11193653 DOI: 10.1016/j.jid.2023.12.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/20/2023] [Accepted: 12/13/2023] [Indexed: 01/16/2024]
Abstract
Cutaneous T-cell lymphomas are mature lymphoid neoplasias resulting from the malignant transformation of skin-resident T-cells. A distinctive clinical feature of cutaneous T-cell lymphomas is their sensitivity to treatment with histone deacetylase inhibitors. However, responses to histone deacetylase inhibitor therapy are universally transient and noncurative, highlighting the need for effective and durable drug combinations. In this study, we demonstrate that the combination of romidepsin, a selective class I histone deacetylase inhibitor, with afatinib, an EGFR family inhibitor, induces strongly synergistic antitumor effects in cutaneous T-cell lymphoma models in vitro and in vivo through abrogation of Jak-signal transducer and activator of transcription signaling. These results support a previously unrecognized potential role for histone deacetylase inhibitor plus afatinib combination in the treatment of cutaneous T-cell lymphomas.
Collapse
Affiliation(s)
- Bobby B Shih
- Institute for Cancer Genetics, Columbia University, New York, New York, USA
| | - Cindy Ma
- Institute for Cancer Genetics, Columbia University, New York, New York, USA
| | - Jose R Cortes
- Institute for Cancer Genetics, Columbia University, New York, New York, USA; Regeneron Pharmaceuticals, Tarrytown, New York, USA
| | - Clara Reglero
- Institute for Cancer Genetics, Columbia University, New York, New York, USA
| | - Hannah Miller
- Institute for Cancer Genetics, Columbia University, New York, New York, USA
| | - S Aidan Quinn
- Institute for Cancer Genetics, Columbia University, New York, New York, USA
| | - Robert Albero
- Institute for Cancer Genetics, Columbia University, New York, New York, USA; Biomedical Research Institute August Pi y Sunyer (IDIBAPS), Barcelona, Spain
| | - Anouchka P Laurent
- Institute for Cancer Genetics, Columbia University, New York, New York, USA
| | - Adam Mackey
- Institute for Cancer Genetics, Columbia University, New York, New York, USA
| | - Adolfo A Ferrando
- Institute for Cancer Genetics, Columbia University, New York, New York, USA; Regeneron Pharmaceuticals, Tarrytown, New York, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA; Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Systems Biology, Columbia University Medical Center, New York, New York, USA
| | - Larisa Geskin
- Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Teresa Palomero
- Institute for Cancer Genetics, Columbia University, New York, New York, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA.
| |
Collapse
|
7
|
Vadivel CK, Willerslev-Olsen A, Namini MRJ, Zeng Z, Yan L, Danielsen M, Gluud M, Pallesen EMH, Wojewoda K, Osmancevic A, Hedebo S, Chang YT, Lindahl LM, Koralov SB, Geskin LJ, Bates SE, Iversen L, Litman T, Bech R, Wobser M, Guenova E, Kamstrup MR, Ødum N, Buus TB. Staphylococcus aureus induces drug resistance in cancer T cells in Sézary syndrome. Blood 2024; 143:1496-1512. [PMID: 38170178 PMCID: PMC11033614 DOI: 10.1182/blood.2023021671] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/16/2023] [Accepted: 12/17/2023] [Indexed: 01/05/2024] Open
Abstract
ABSTRACT Patients with Sézary syndrome (SS), a leukemic variant of cutaneous T-cell lymphoma (CTCL), are prone to Staphylococcus aureus infections and have a poor prognosis due to treatment resistance. Here, we report that S aureus and staphylococcal enterotoxins (SE) induce drug resistance in malignant T cells against therapeutics commonly used in CTCL. Supernatant from patient-derived, SE-producing S aureus and recombinant SE significantly inhibit cell death induced by histone deacetylase (HDAC) inhibitor romidepsin in primary malignant T cells from patients with SS. Bacterial killing by engineered, bacteriophage-derived, S aureus-specific endolysin (XZ.700) abrogates the effect of S aureus supernatant. Similarly, mutations in major histocompatibility complex (MHC) class II binding sites of SE type A (SEA) and anti-SEA antibody block induction of resistance. Importantly, SE also triggers resistance to other HDAC inhibitors (vorinostat and resminostat) and chemotherapeutic drugs (doxorubicin and etoposide). Multimodal single-cell sequencing indicates T-cell receptor (TCR), NF-κB, and JAK/STAT signaling pathways (previously associated with drug resistance) as putative mediators of SE-induced drug resistance. In support, inhibition of TCR-signaling and Protein kinase C (upstream of NF-κB) counteracts SE-induced rescue from drug-induced cell death. Inversely, SE cannot rescue from cell death induced by the proteasome/NF-κB inhibitor bortezomib. Inhibition of JAK/STAT only blocks rescue in patients whose malignant T-cell survival is dependent on SE-induced cytokines, suggesting 2 distinct ways SE can induce drug resistance. In conclusion, we show that S aureus enterotoxins induce drug resistance in primary malignant T cells. These findings suggest that S aureus enterotoxins cause clinical treatment resistance in patients with SS, and antibacterial measures may improve the outcome of cancer-directed therapy in patients harboring S aureus.
Collapse
Affiliation(s)
- Chella Krishna Vadivel
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Willerslev-Olsen
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Martin R. J. Namini
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Ziao Zeng
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Lang Yan
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Maria Danielsen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Maria Gluud
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Emil M. H. Pallesen
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Karolina Wojewoda
- Department of Dermatology and Venereology, Region Västra Götaland, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Amra Osmancevic
- Department of Dermatology and Venereology, Region Västra Götaland, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Signe Hedebo
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Yun-Tsan Chang
- Department of Dermatology and Venereology, University Hospital Centre (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Lise M. Lindahl
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Sergei B. Koralov
- Department of Pathology, New York University School of Medicine, New York, NY
| | - Larisa J. Geskin
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY
| | - Susan E. Bates
- Division of Hematology/Oncology, Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY
| | - Lars Iversen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas Litman
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Rikke Bech
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Marion Wobser
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Emmanuella Guenova
- Department of Dermatology and Venereology, University Hospital Centre (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Maria R. Kamstrup
- Department of Dermatology, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Niels Ødum
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Terkild B. Buus
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Fay CJ, Awh KC, LeBoeuf NR, Larocca CA. Harnessing the immune system in the treatment of cutaneous T cell lymphomas. Front Oncol 2023; 12:1071171. [PMID: 36713518 PMCID: PMC9878398 DOI: 10.3389/fonc.2022.1071171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/01/2022] [Indexed: 01/15/2023] Open
Abstract
Cutaneous T cell lymphomas are a rare subset of non-Hodgkin's lymphomas with predilection for the skin with immunosuppressive effects that drive morbidity and mortality. We are now appreciating that suppression of the immune system is an important step in the progression of disease. It should come as no surprise that therapies historically and currently being used to treat these cancers have immune modulating functions that impact disease outcomes. By understanding the immune effects of our therapies, we may better develop new agents that target the immune system and improve combinatorial treatment strategies to limit morbidity and mortality of these cancers. The immune modulating effect of therapeutic drugs in use and under development for cutaneous T cell lymphomas will be reviewed.
Collapse
|
9
|
Qu H, Wang M, Wang M, Liu Y, Quan C. The expression and the tumor suppressor role of CLDN6 in colon cancer. Mol Cell Biochem 2022; 477:2883-2893. [PMID: 35701678 DOI: 10.1007/s11010-022-04450-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/24/2022] [Indexed: 11/29/2022]
Abstract
As a member of the tight junction family, CLDN6 is a tumor suppressor in breast cancer, but its role in colon cancer is unknown. In this research, we aimed at revealing the function of CLDN6 in colon cancer. We found that colon cancer tissues lowly expressed CLDN6, and the expression of CLDN6 was negatively correlated with lymph node metastasis. Similarly, CLDN6 was lowly expressed in the colon cancer cell line SW1116, and overexpression of CLDN6 inhibited cell proliferation in vitro and in vivo. Consistently, the migration and invasion abilities of cells were significantly inhibited after CLDN6 overexpression. In addition, we demonstrated that CLDN6 may inhibit the migration and invasion abilities by activating the TYK2/STAT3 pathway. Therefore, our data indicated that CLDN6 acted as a tumor suppressor and had the potential to be regarded as a biomarker for the progression of colon cancer.
Collapse
Affiliation(s)
- Huinan Qu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, People's Republic of China
| | - Min Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, People's Republic of China
| | - Miaomiao Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, People's Republic of China
| | - Yuanyuan Liu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, People's Republic of China
| | - Chengshi Quan
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, People's Republic of China.
| |
Collapse
|
10
|
Kołkowski K, Trzeciak M, Sokołowska-Wojdyło M. Safety and Danger Considerations of Novel Treatments for Atopic Dermatitis in Context of Primary Cutaneous Lymphomas. Int J Mol Sci 2021; 22:13388. [PMID: 34948183 PMCID: PMC8703592 DOI: 10.3390/ijms222413388] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 12/24/2022] Open
Abstract
The impact of new and emerging therapies on the microenvironment of primary cutaneous lymphomas (PCLs) has been recently raised in the literature. Concomitantly, novel treatments are already used or registered (dupilumab, upadacitinib) and others seem to be added to the armamentarium against atopic dermatitis. Our aim was to review the literature on interleukins 4, 13, 22, and 31, and JAK/STAT pathways in PCLs to elucidate the safety of using biologics (dupilumab, tralokinumab, fezakinumab, nemolizumab) and small molecule inhibitors (upadacitinib, baricitinib, abrocitinib, ruxolitinib, tofacitinib) in the treatment of atopic dermatitis. We summarized the current state of knowledge on this topic based on the search of the PubMed database and related references published before 21 October 2021. Our analysis suggests that some of the mentioned agents (dupilumab, ruxolitinib) and others may have a direct impact on the progression of cutaneous lymphomas. This issue requires further study and meticulous monitoring of patients receiving these drugs to ensure their safety, especially in light of the FDA warning on tofacitinib. In conclusion, in the case of the rapid progression of atopic dermatitis/eczema, especially in patients older than 40 years old, there is a necessity to perform a biopsy followed by a very careful pathological examination.
Collapse
Affiliation(s)
- Karol Kołkowski
- Dermatological Students Scientific Association, Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland
| | - Magdalena Trzeciak
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland; (M.T.); (M.S.-W.)
| | - Małgorzata Sokołowska-Wojdyło
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland; (M.T.); (M.S.-W.)
| |
Collapse
|
11
|
Yang Y, Liu J, Kamounah FS, Ciancaleoni G, Lee JW. A CO 2-Catalyzed Transamidation Reaction. J Org Chem 2021; 86:16867-16881. [PMID: 34723529 DOI: 10.1021/acs.joc.1c02077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Transamidation reactions are often mediated by reactive substrates in the presence of overstoichiometric activating reagents and/or transition metal catalysts. Here we report the use of CO2 as a traceless catalyst: in the presence of catalytic amounts of CO2, transamidation reactions were accelerated with primary, secondary, and tertiary amide donors. Various amine nucleophiles including amino acid derivatives were tolerated, showcasing the utility of transamidation in peptide modification and polymer degradation (e.g., Nylon-6,6). In particular, N,O-dimethylhydroxyl amides (Weinreb amides) displayed a distinct reactivity in the CO2-catalyzed transamidation versus a N2 atmosphere. Comparative Hammett studies and kinetic analysis were conducted to elucidate the catalytic activation mechanism of molecular CO2, which was supported by DFT calculations. We attributed the positive effect of CO2 in the transamidation reaction to the stabilization of tetrahedral intermediates by covalent binding to the electrophilic CO2.
Collapse
Affiliation(s)
- Yang Yang
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, Copenhagen Ø 2100, Denmark
| | - Jian Liu
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, Copenhagen Ø 2100, Denmark
| | - Fadhil S Kamounah
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, Copenhagen Ø 2100, Denmark
| | - Gianluca Ciancaleoni
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy.,CIRCC, via Celso Ulpiani 27, I-70126 Bari, Italy
| | - Ji-Woong Lee
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, Copenhagen Ø 2100, Denmark.,Nanoscience Center, University of Copenhagen, Universitetsparken 5, Copenhagen Ø 2100, Denmark
| |
Collapse
|
12
|
Xiao MZX, Hennessey D, Iyer A, O'Keefe S, Zhang F, Sivanand A, Gniadecki R. Transcriptomic Changes During Stage Progression of Mycosis Fungoides. Br J Dermatol 2021; 186:520-531. [PMID: 34528236 DOI: 10.1111/bjd.20760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Mycosis fungoides (MF) is the most common cutaneous T cell lymphoma, which in the early patch/plaque stages runs an indolent course. However, ~25% of MF patients develop skin tumors, a hallmark of progression to the advanced stage and is associated with high mortality. The mechanisms involved in stage progression are poorly elucidated. METHODS We performed whole-transcriptome and whole-exome sequencing of malignant MF cells from skin biopsies obtained by laser-capture microdissection. We compared three types of MF lesions: early-stage plaques (ESP, n=12) as well as plaques and tumors from patients in late-stage disease (late-stage plaques [LSP], n=10, and tumors [TMR], n=15). Gene Ontology (GO) and KEGG analysis were used to determine pathway changes specific for different lesions which were linked to the recurrent somatic mutations overrepresented in MF tumors. RESULTS The key upregulated pathways during stage progression were those related to cell proliferation and survival (MEK/ERK, Akt-mTOR), Th2/Th9 signaling (IL4, STAT3, STAT5, STAT6), meiomitosis (CT45A1, CT45A3, STAG3, GTSF1, REC8) and DNA repair (PARP1, MYCN, OGG1). Principal coordinate clustering of the transcriptome revealed extensive gene expression differences between early (ESP) and advanced-stage lesions (LSP and TMR). LSP and TMR showed remarkable similarities at the level of the transcriptome, which we interpreted as evidence of cell percolation between lesions via hematogenous self-seeding. CONCLUSION Stage progression in MF is associated with Th2/Th9 polarization of malignant cells, activation of proliferation, survival, as well as increased genomic instability. Global transcriptomic changes in multiple lesions may be caused by hematogenous cell percolation between discrete skin lesions.
Collapse
Affiliation(s)
- M Z X Xiao
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| | - D Hennessey
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| | - A Iyer
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| | - S O'Keefe
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| | - F Zhang
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - A Sivanand
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| | - R Gniadecki
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
13
|
Willerslev-Olsen A, Gjerdrum LMR, Lindahl LM, Buus TB, Pallesen EMH, Gluud M, Bzorek M, Nielsen BS, Kamstrup MR, Rittig AH, Bonefeld CM, Krejsgaard T, Geisler C, Koralov SB, Litman T, Becker JC, Woetmann A, Iversen L, Odum N. Staphylococcus aureus Induces Signal Transducer and Activator of Transcription 5‒Dependent miR-155 Expression in Cutaneous T-Cell Lymphoma. J Invest Dermatol 2021; 141:2449-2458. [PMID: 33862068 DOI: 10.1016/j.jid.2021.01.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/26/2022]
Abstract
Staphylococcal enterotoxins are believed to fuel disease activity in cutaneous T-cell lymphoma. Recent data support this by showing that antibiotics inhibit malignant T cells in skin lesions in mycosis fungoides and Sézary syndrome, the most common forms of cutaneous T-cell lymphoma. Yet, it remains incompletely characterized how staphylococcal enterotoxins fuel disease activity. In this study, we show that staphylococcal enterotoxins induce the expression of the oncogenic microRNA miR-155 in primary malignant T cells. Thus, staphylococcal enterotoxins and Staphyloccocus aureus isolates from lesional skin of patients induce miR-155 expression at least partly through the IL-2Rg‒Jak‒signal transducer and activator of transcription 5 pathway, and the effect is augmented by the presence of nonmalignant T cells. Importantly, mycosis fungoides lesions harbor S. aureus, express Y-phosphorylated signal transducer and activator of transcription 5, and display enhanced miR-155 expression, when compared with nonlesional and healthy skin. Preliminary data show that aggressive antibiotic therapy is associated with decreased Y-phosphorylated signal transducer and activator of transcription 5 and miR-155 expression in lesional skin in two patients with Sézary syndrome. In conclusion, we show that S. aureus and its enterotoxins induce enhanced expression of oncogenic miR-155, providing mechanistic insight into the role of S. aureus in cutaneous T-cell lymphoma. Our findings support that environmental stimuli such as bacteria can fuel disease progression in cutaneous T-cell lymphoma.
Collapse
Affiliation(s)
- Andreas Willerslev-Olsen
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Lise Mette Rahbek Gjerdrum
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Lise M Lindahl
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Terkild B Buus
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Emil M H Pallesen
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Maria Gluud
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Michael Bzorek
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | | | - Maria R Kamstrup
- Department of Dermatology, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Anne Hald Rittig
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Charlotte M Bonefeld
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Thorbjørn Krejsgaard
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Geisler
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Sergei B Koralov
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Thomas Litman
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jurgen C Becker
- Department of Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Hospital of Essen, Essen, Germany; Deutsches Krebsforschungsinstitut (DKFZ), Heidelberg, Germany
| | - Anders Woetmann
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Lars Iversen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Niels Odum
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
14
|
Ou-Yang H, Wu SC, Sung LY, Yang SH, Yang SH, Chong KY, Chen CM. STAT3 Is an Upstream Regulator of Granzyme G in the Maternal-To-Zygotic Transition of Mouse Embryos. Int J Mol Sci 2021; 22:ijms22010460. [PMID: 33466434 PMCID: PMC7796490 DOI: 10.3390/ijms22010460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/19/2020] [Accepted: 12/31/2020] [Indexed: 12/24/2022] Open
Abstract
The maternal-to-zygotic transition (MZT), which controls maternal signaling to synthesize zygotic gene products, promotes the preimplantation development of mouse zygotes to the two-cell stage. Our previous study reported that mouse granzyme g (Gzmg), a serine-type protease, is required for the MZT. In this study, we further identified the maternal factors that regulate the Gzmg promoter activity in the zygote to the two-cell stage of mouse embryos. A full-length Gzmg promoter from mouse genomic DNA, FL-pGzmg (−1696~+28 nt), was cloned, and four deletion constructs of this Gzmg promoter, Δ1-pGzmg (−1369~+28 nt), Δ2-pGzmg (−939~+28 nt), Δ3-pGzmg (−711~+28 nt) and Δ4-pGzmg (−417~+28 nt), were subsequently generated. Different-sized Gzmg promoters were used to perform promoter assays of mouse zygotes and two-cell stage embryos. The results showed that Δ4-pGzmg promoted the highest expression level of the enhanced green fluorescent protein (EGFP) reporter in the zygotes and two-cell embryos. The data suggested that time-specific transcription factors upregulated Gzmg by binding cis-elements in the −417~+28-nt Gzmg promoter region. According to the results of the promoter assay, the transcription factor binding sites were predicted and analyzed with the JASPAR database, and two transcription factors, signal transducer and activator of transcription 3 (STAT3) and GA-binding protein alpha (GABPα), were identified. Furthermore, STAT3 and GABPα are expressed and located in zygote pronuclei and two-cell nuclei were confirmed by immunofluorescence staining; however, only STAT3 was recruited to the mouse zygote pronuclei and two-cell nuclei injected with the Δ4-pGzmg reporter construct. These data indicated that STAT3 is a maternal transcription factor and may upregulate Gzmg to promote the MZT. Furthermore, treatment with a STAT3 inhibitor, S3I-201, caused mouse embryonic arrest at the zygote and two-cell stages. These results suggest that STAT3, a maternal protein, is a critical transcription factor and regulates Gzmg transcription activity in preimplantation mouse embryos. It plays an important role in the maternal-to-zygotic transition during early embryonic development.
Collapse
Affiliation(s)
- Huan Ou-Yang
- Department of Life Sciences, and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (H.O.-Y.); (S.-H.Y.)
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan;
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan;
| | - Shinn-Chih Wu
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan;
| | - Li-Ying Sung
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan;
| | - Shiao-Hsuan Yang
- Department of Life Sciences, and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (H.O.-Y.); (S.-H.Y.)
- Reproductive Medicine Center, Department of Gynecology, Changhua Christian Hospital, Changhua 515, Taiwan
| | - Shang-Hsun Yang
- Department of Physiology, National Cheng Kung University, Tainan 70101, Taiwan;
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan
| | - Kowit-Yu Chong
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (H.O.-Y.); (S.-H.Y.)
- The iEGG and Animal Biotechnology Center, and Rong-Hsing Translational Medicine Research Center, National Chung Hsing University, Taichung 402, Taiwan
- Correspondence: ; Tel.: +886-4-22856309
| |
Collapse
|
15
|
Fan L, Zhou L. AG490 protects cerebral ischemia/reperfusion injury via inhibiting the JAK2/3 signaling pathway. Brain Behav 2021; 11:e01911. [PMID: 33098244 PMCID: PMC7821583 DOI: 10.1002/brb3.1911] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cerebral ischemia/reperfusion injury is a severe problem in patients with brain ischemia. Brain injury caused by the immune response is important in the pathogenesis of cerebral ischemia/reperfusion injury and immune pathways. It is important to investigate potential targets for the treatment of cerebral ischemia/reperfusion injury. METHODS In this experiment, we evaluated the effect of an exogenous JAK antagonist AG490 in the cerebral ischemia/reperfusion injury model, which was established by middle cerebral artery occlusion (MCAO). Histology study, TUNEL staining, Western blot, and RT-PCR were employed to examine the effects of AG490 in cerebral ischemia/reperfusion injury. RESULTS In the brain tissue of MCAO mice, JAK2 was highly expressed. AG490 is an inhibitor of JAK2, which reduced the phosphorylation level of JAK2. AG490 downregulated the phosphorylated activation of JAK3 and their downstream STAT3. The antiapoptotic activity of AG490 on cerebral ischemia/reperfusion injury mice was consistent with in vitro data. It reduced the phosphorylation of JAK2/JAK3/STAT3 and the apoptosis rate in cultured neurons upon apoptosis induction. Besides, we also observed the neuroprotective effects of AG490 on cerebral ischemia/reperfusion injury. Administration of AG490 could further enhance the expression of neurotrophins including BNDF, NT3, and the neurotrophin receptor TrkB. CONCLUSION Therefore, AG490 is pluripotent for cerebral ischemia/reperfusion injury through both antiapoptosis and neuroprotective activities. The antiapoptosis effect is dependent on its regulation of the JAK-STAT pathway.
Collapse
Affiliation(s)
- Lichao Fan
- Department of Neurology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Lichun Zhou
- Department of Neurology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Liu Y, Liao S, Bennett S, Tang H, Song D, Wood D, Zhan X, Xu J. STAT3 and its targeting inhibitors in osteosarcoma. Cell Prolif 2020; 54:e12974. [PMID: 33382511 PMCID: PMC7848963 DOI: 10.1111/cpr.12974] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/21/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is one of seven STAT family members involved with the regulation of cellular growth, differentiation and survival. STAT proteins are conserved among eukaryotes and are important for biological functions of embryogenesis, immunity, haematopoiesis and cell migration. STAT3 is widely expressed and located in the cytoplasm in an inactive form. STAT3 is rapidly and transiently activated by tyrosine phosphorylation by a range of signalling pathways, including cytokines from the IL‐6 family and growth factors, such as EGF and PDGF. STAT3 activation and subsequent dimer formation initiates nuclear translocation of STAT3 for the regulation of target gene transcription. Four STAT3 isoforms have been identified, which have distinct biological functions. STAT3 is considered a proto‐oncogene and constitutive activation of STAT3 is implicated in the development of various cancers, including multiple myeloma, leukaemia and lymphomas. In this review, we focus on recent progress on STAT3 and osteosarcoma (OS). Notably, STAT3 is overexpressed and associated with the poor prognosis of OS. Constitutive activation of STAT3 in OS appears to upregulate the expression of target oncogenes, leading to OS cell transformation, proliferation, tumour formation, invasion, metastasis, immune evasion and drug resistance. Taken together, STAT3 is a target for cancer therapy, and STAT3 inhibitors represent potential therapeutic candidates for the treatment of OS.
Collapse
Affiliation(s)
- Yun Liu
- Department of Spine and Osteopathic Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.,Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia.,Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Shijie Liao
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia.,Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China.,Department of Trauma Orthopedic and Hand Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Samuel Bennett
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Haijun Tang
- Department of Orthopedic, Guangxi hospital for nationalities, Nanning, Guangxi, China
| | - Dezhi Song
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia.,Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - David Wood
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Xinli Zhan
- Department of Spine and Osteopathic Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.,Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia.,Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiake Xu
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
17
|
Epigenetic Silencing of Tumor Suppressor miR-124 Directly Supports STAT3 Activation in Cutaneous T-Cell Lymphoma. Cells 2020; 9:cells9122692. [PMID: 33333886 PMCID: PMC7765332 DOI: 10.3390/cells9122692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence supports a potential role for STAT3 as a tumor driver in cutaneous T-cell lymphomas (CTCL). The mechanisms leading to STAT3 activation are not fully understood; however, we recently found that miR-124, a known STAT3 regulator, is robustly silenced in MF tumor-stage and CTCL cells. Objective: We studied here whether deregulation of miR-124 contributes to STAT3 pathway activation in CTCL. Methods: We measured the effect of ectopic mir-124 expression in active phosphorylated STAT3 (p-STAT3) levels and evaluated the transcriptional impact of miR-124-dependent STAT3 pathway regulation by expression microarray analysis. Results: We found that ectopic expression of miR-124 results in massive downregulation of activated STAT3 in different CTCL lines, which resulted in a significant alteration of genetic signatures related with gene transcription and proliferation such as MYC and E2F. Conclusions: Our study highlights the importance of the miR-124/STAT3 axis in CTCL and demonstrates that the STAT3 pathway is regulated through epigenetic mechanisms in these cells. Since deregulated STAT3 signaling has a major impact on CTCL initiation and progression, a better understanding of the molecular basis of the miR-124/STAT3 axis may provide useful information for future personalized therapies.
Collapse
|
18
|
STAT3 signaling pathway is involved in the pathogenesis of miscarriage. Placenta 2020; 101:30-38. [DOI: 10.1016/j.placenta.2020.08.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 08/18/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022]
|
19
|
Busker S, Page B, Arnér ESJ. To inhibit TrxR1 is to inactivate STAT3-Inhibition of TrxR1 enzymatic function by STAT3 small molecule inhibitors. Redox Biol 2020; 36:101646. [PMID: 32863208 PMCID: PMC7378686 DOI: 10.1016/j.redox.2020.101646] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/03/2020] [Accepted: 07/11/2020] [Indexed: 01/05/2023] Open
Abstract
The transcription factor STAT3 plays a key role in cancer and immunity, being widely explored as a potential drug target for the development of novel immunomodulatory or anticancer therapeutics. The mechanisms of small molecule-derived inhibition of STAT3 appear, however, to be more complex than initially perceived. Our recent discovery, that some novel STAT3 inhibitors were bona fide inhibitors of the cytosolic selenoprotein oxidoreductase TrxR1 (TXNRD1), led us to explore the effects of a wide array of previously described STAT3 inhibitors on TrxR1 function. We found that 17 out of 23 tested STAT3 small molecule inhibitors indeed inhibited purified TrxR1 at the reported concentrations yielding STAT3 inhibition. All tested compounds were electrophilic as shown by direct reactivities with GSH, and several were found to also be redox cycling substrates of TrxR1. Ten compounds previously shown to inhibit STAT3 were here found to irreversibly inhibit cellular TrxR1 activity (Auranofin, Stattic, 5,15-DPP, Galiellalactone, LLL12, Napabucasin, BP1-102, STA-21, S3I-201 and Degrasyn (WP1130)). Our findings suggest that targeting of TrxR1 may be a common feature for many small molecules that inhibit cellular STAT3 function. It is possible that prevention of STAT3 activation in cells by several small molecules classified as STAT3 inhibitors can be a downstream event following TrxR1 inhibition. Therefore, the relationship between TrxR1 and STAT3 should be considered when studying inhibition of either of these promising drug targets.
Collapse
Affiliation(s)
- Sander Busker
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Brent Page
- Department of Oncology-Pathology, Science for Life Laboratories, Karolinska Institutet, Stockholm, Sweden; Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; Department of Selenoprotein Research, National Institute of Oncology, Budapest, Hungary.
| |
Collapse
|
20
|
Trager MH, Geskin LJ. Current status of histone deacetylase inhibitors in cutaneous T-cell lymphoma. GIORN ITAL DERMAT V 2020; 154:681-695. [PMID: 31859467 DOI: 10.23736/s0392-0488.19.06503-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cutaneous T cell lymphoma (CTCL) is a non-Hodgkin's lymphoma with a heterogenous presentation and highly variable disease course. The most common subtypes of CTCL are mycosis fungoides (MF) and Sézary Syndrome (SS). Treatment varies based on the stage of the disease with skin directed therapies typically utilized for early stage disease, and systemic therapies employed for more advanced disease. There are few highly effective treatments available, and systemic therapies have limited response rates. Histone deacetylase inhibitors have emerged as mainstream treatments for MF/SS over the past several years. Here, we discuss the mechanism of action of histone deacetylase inhibitors in relation to the pathogenesis of MF/SS, evaluate the clinical trials that led to Food and Drug Administration approval of two of the histone deacetylase inhibitors for MF/SS and describe the results for those still under investigation. Additionally, we discuss the potential for combination therapies in order to optimize outcomes of treatment with histone deacetylase inhibitors.
Collapse
Affiliation(s)
- Megan H Trager
- Columbia University, Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Larisa J Geskin
- Department of Dermatology, Irving Medical Center, Columbia University, New York, NY, USA -
| |
Collapse
|
21
|
Zhou Y, Sun Y, Hou W, Ma L, Tao Y, Li D, Xu C, Bao J, Fan W. The JAK2/STAT3 pathway inhibitor, AG490, suppresses the abnormal behavior of keloid fibroblasts in vitro. Int J Mol Med 2020; 46:191-200. [PMID: 32377718 PMCID: PMC7255460 DOI: 10.3892/ijmm.2020.4592] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/09/2020] [Indexed: 01/01/2023] Open
Abstract
AG490 is a selective inhibitor of the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway. The present study examined its effects on the abnormal behavior of human keloid fibroblasts (HKFs) and evaluated its potential use in the treatment of keloids. Human normal fibroblasts (HNFs) and HKFs were treated with increasing concentrations of AG490. The proliferation of HNFs and HKFs was inhibited by AG490 in both a time‑ and concentration‑dependent manner by increasing apoptosis and inducing G1 cell cycle arrest. The downregulation of cyclin D1 and connective tissue growth factor (CTGF) expression was associated with a decrease in STAT3 expression in response to AG490. The effects of AG490 on TGF‑β‑stimulated fibroblasts, including HNFs, HKFs and hypertrophic scar fibroblasts (HSFs) were also evaluated. The TGF‑β1‑stimulated excessive proliferation and CTGF production were markedly inhibited by the application of AG490 in the HNFs, HSFs and HKFs. In addition, the STAT3‑specific decoy oligodeoxynucleotides (SODNs) were transfected into HKFs. The invasive ability of the SODN‑transfected HKFs was determined and the expression of extracellular matrix components was quantified. Similarly, SODNs blocked the constitutive activation of STAT3. SODNs inhibited the invasion and progression of HKFs, possibly via the upregulation of the expression of tissue inhibitor of metalloproteinase‑2 (TIMP‑2), and the downregulation of the expression of matrix metalloproteinase‑2 (MMP‑2) and vascular endothelial growth factor (VEGF). On the whole, the findings of the present study demonstrate that STAT3‑specific elimination, such as the application of AG490 and decoy ODNs, may serve as promising therapeutic strategies for the treatment of keloids.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Dermatology, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China
| | - Yuexin Sun
- Department of Dermatology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, P.R. China
| | - Wenjun Hou
- Department of Dermatology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, P.R. China
| | - Liwen Ma
- Department of Dermatology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, P.R. China
| | - Yue Tao
- Department of Dermatology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, P.R. China
| | - Dan Li
- Department of Dermatology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, P.R. China
| | - Cui Xu
- Department of Dermatology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, P.R. China
| | - Jun Bao
- Department of Dermatology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu 210008, P.R. China
| | - Weixin Fan
- Department of Dermatology, Jiangsu Province People's Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
22
|
The polymorphisms of IL-6/STAT3 signaling pathway may contribute to cutaneous T-cell lymphomas susceptibility. Arch Dermatol Res 2020; 313:25-31. [PMID: 32270320 PMCID: PMC7806529 DOI: 10.1007/s00403-020-02062-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 01/26/2020] [Accepted: 03/28/2020] [Indexed: 12/15/2022]
Abstract
IL-6/STAT3 signaling pathway has been suggested to play a role in CTCL pathogenesis. Polymorphisms in STAT3 signaling pathway-related genes might be a risk factor for CTCL. However, the exact role of inherited gene polymorphisms of IL-6 and STAT3 in the pathogenesis of CTCL is still not fully understood. The aim was to examine whether IL-6 cytokine and polymorphisms of IL-6 and STAT3 gene are associated with CTCL susceptibility, stage of disease and pruritus intensity. We compared the IL-6 serum level and the frequency of selected single nucleotide polymorphisms of IL-6 and STAT3 in 106 CTCL and 198 control group using polymerase chain reaction with sequence-specific primers method and ELISA. We have found that serum IL-6 level in CTCL patients was significantly higher than in healthy controls (p < 0.05). We also demonstrated that two genotypes, CC of IL-6 and GG of STAT3, were overexpressed in CTCL patients compared to healthy controls, and that they increase the risk of malignancy development (OR = 1.8, p = 0.04 for IL-6 and OR 2.53, p = 0.0064 for STAT3). Moreover, the GG genotype of STAT3 polymorphism seems to be associated with lack of pruritus or mild pruritus in CTCL patients. Our results indicate that IL-6 is involved in pathogenesis of CTCL but not pruritus. Moreover, CC of IL-6 and GG genotype of STAT3 genes might be considered as the risk factor for development of CTCL.
Collapse
|
23
|
STAT3 Dysregulation in Mature T and NK Cell Lymphomas. Cancers (Basel) 2019; 11:cancers11111711. [PMID: 31684088 PMCID: PMC6896161 DOI: 10.3390/cancers11111711] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 10/25/2019] [Accepted: 10/29/2019] [Indexed: 12/21/2022] Open
Abstract
Abstract: T cell lymphomas comprise a distinct class of non-Hodgkin's lymphomas, which include mature T and natural killer (NK) cell neoplasms. While each malignancy within this group is characterized by unique clinicopathologic features, dysregulation in the Janus tyrosine family of kinases/Signal transducer and activator of transcription (JAK/STAT) signaling pathway, specifically aberrant STAT3 activation, is a common feature among these lymphomas. The mechanisms driving dysregulation vary among T cell lymphoma subtypes and include activating mutations in upstream kinases or STAT3 itself, formation of oncogenic kinases which drive STAT3 activation, loss of negative regulators of STAT3, and the induction of a pro-tumorigenic inflammatory microenvironment. Constitutive STAT3 activation has been associated with the expression of targets able to increase pro-survival signals and provide malignant fitness. Patients with dysregulated STAT3 signaling tend to have inferior clinical outcomes, which underscores the importance of STAT3 signaling in malignant progression. Targeting of STAT3 has shown promising results in pre-clinical studies in T cell lymphoma lines, ex-vivo primary malignant patient cells, and in mouse models of disease. However, targeting this pleotropic pathway in patients has proven difficult. Here we review the recent contributions to our understanding of the role of STAT3 in T cell lymphomagenesis, mechanisms driving STAT3 activation in T cell lymphomas, and current efforts at targeting STAT3 signaling in T cell malignancies.
Collapse
|
24
|
Pérez C, Mondéjar R, García-Díaz N, Cereceda L, León A, Montes S, Durán Vian C, Pérez Paredes MG, González-Morán A, Alegre de Miguel V, Sanz Anquela JM, Frias J, Limeres MA, González LM, Martín Dávila F, Beltrán M, Mollejo M, Méndez JR, González MA, González García J, López R, Gómez A, Izquierdo F, Ramos R, Camacho C, Rodriguez-Pinilla SM, Martínez N, Vaqué JP, Ortiz-Romero PL, Piris MA. Advanced-stage mycosis fungoides: role of the signal transducer and activator of transcription 3, nuclear factor-κB and nuclear factor of activated T cells pathways. Br J Dermatol 2019; 182:147-155. [PMID: 31049933 DOI: 10.1111/bjd.18098] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND The malignant mechanisms that control the development of cutaneous T-cell lymphoma (CTCL) are beginning to be identified. Recent evidence suggests that disturbances in specific intracellular signalling pathways, such as RAS-mitogen-activated protein kinase, T-cell receptor (TCR)-phospholipase C gamma 1 (PLCG1)-nuclear factor of activated T cells (NFAT) and Janus kinase (JAK)-signal transducer and activator of transcription (STAT), may play an essential role in the pathogenesis of CTCL. OBJECTIVES To investigate the mechanisms controlling disease development and progression in mycosis fungoides (MF), the most common form of CTCL. METHODS We collected 100 samples that were submitted for diagnosis of, or a second opinion regarding, MF between 2001 and 2018, 80% of which were in the early clinical stages of the disease. Formalin-fixed paraffin-embedded tissues were used for histological review and to measure the expression by immunohistochemistry of surrogate markers of activation of the TCR-PLCG1-NFAT, JAK-STAT and NF-κB pathways. Folliculotropism and large-cell transformation were also examined. RESULTS NFAT and nuclear factor kappa B (NF-κB) markers showed a comparable activation status in early and advanced stages, while STAT3 activation was more frequent in advanced stages and was associated with large-cell transformation. Consistently with this observation, STAT3 activation occurred in parallel with MF progression in two initially MF-negative cases. A significant association of NFAT with NF-κB markers was also found, reflecting a common mechanism of activation in the two pathways. Genomic studies identified nine mutations in seven genes known to play a potential role in tumorigenesis in T-cell leukaemia/lymphoma, including PLCG1, JAK3 and STAT3, which underlies the activation of these key cell-survival pathways. A higher mutational allele frequency was detected in advanced stages. CONCLUSIONS Our results show that STAT3 is activated in advanced cases and is associated with large-cell transformation, while the activation of NFAT and NF-κB is maintained throughout the disease. These findings could have important diagnostic and therapeutic implications. What's already known about this topic? Mycosis fungoides is characterized by a clonal expansion of T cells in the skin. The mechanisms controlling disease development and progression are not fully understood. What does this study add? An association of the nuclear factor of activated T cells and nuclear factor kappa B pathways was found, which could reflect a common mechanism of activation. These pathways were activated in early and advanced stages at the same level. Signal transducer and activator of transcription 3 activation was associated with large-cell transformation and was more frequent in advanced stages. A genomic analysis of cutaneous T-cell lymphoma-associated genes was performed. Nine mutations were detected. What is the translational message? These results could have important implications for the treatment of MF in the near future.
Collapse
Affiliation(s)
- C Pérez
- Translational Hematopathology, Instituto de Investigación Marqués de Valdecilla, IDIVAL, Santander, Spain.,Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - R Mondéjar
- Translational Hematopathology, Instituto de Investigación Marqués de Valdecilla, IDIVAL, Santander, Spain.,Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain.,Service of Pathology, Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - N García-Díaz
- Departamento de Biología Molecular, Universidad de Cantabria, Infección, Inmunidad y Patología Digestive, Instituto de Investigación Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - L Cereceda
- Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain.,Service of Pathology, Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - A León
- Pathology Service, Marqués de Valdecilla University Hospital, Santander, Spain
| | - S Montes
- Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain.,Pathology Service, Marqués de Valdecilla University Hospital, Santander, Spain
| | - C Durán Vian
- Dermatology Service, Marqués de Valdecilla University Hospital, Santander, Spain
| | - M G Pérez Paredes
- Dermatology Service, Marqués de Valdecilla University Hospital, Santander, Spain
| | - A González-Morán
- Dermatology Service, Complejo Hospitalario de Ávila, Ávila, Spain
| | - V Alegre de Miguel
- Dermatology Service, Hospital General Universitario de Valencia, Valencia, Spain
| | - J M Sanz Anquela
- Cancer Registry and Pathology Department, Hospital Universitario Príncipe de Asturias and Department of Medicine and Medical Specialties, Faculty of Medicine, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - J Frias
- Dermatology Service, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - M A Limeres
- Pathology Department, Hospital Universitario de Gran Canaria Doctor Negrín, Gran Canaria, Canarias, Spain
| | - L M González
- Pathology Service, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - F Martín Dávila
- Pathology Service, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - M Beltrán
- Pathology Service, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - M Mollejo
- Pathology Service, Complejo Hospitalario de Toledo, Toledo, Spain
| | - J R Méndez
- Pathology Service, Centro Médico de Asturias, Asturias, Spain
| | - M A González
- Pathology Service, Hospital San Pedro de Alcántara, Cáceres, Spain
| | - J González García
- Pathology Service, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - R López
- Pathology Service, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - A Gómez
- Pathology Service, Hospital de la Marina Baixa, Alicante, Spain
| | - F Izquierdo
- Pathology Service, Complejo Asistencial Universitario de León, León, Spain
| | - R Ramos
- Pathology Service, University Hospital Son Espases, Palma de Mallorca, Spain
| | - C Camacho
- Pathology Service, C.H.U. Insular - Materno Infantil, Gran Canarias, Spain
| | - S M Rodriguez-Pinilla
- Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain.,Service of Pathology, Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - N Martínez
- Translational Hematopathology, Instituto de Investigación Marqués de Valdecilla, IDIVAL, Santander, Spain.,Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - J P Vaqué
- Departamento de Biología Molecular, Universidad de Cantabria, Infección, Inmunidad y Patología Digestive, Instituto de Investigación Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - P L Ortiz-Romero
- Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain.,Dermatology Service, Hospital 12 de Octubre, Institute i+12 Medical School, University Complutense, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| | - M A Piris
- Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain.,Service of Pathology, Fundación Jiménez Díaz University Hospital, Madrid, Spain
| |
Collapse
|
25
|
Gug G, Huang Q, Chiticariu E, Solovan C, Baudis M. DNA copy number imbalances in primary cutaneous lymphomas. J Eur Acad Dermatol Venereol 2019; 33:1062-1075. [PMID: 30659659 DOI: 10.1111/jdv.15442] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/14/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Cutaneous lymphomas (CL) represent a clinically defined group of extranodal non-Hodgkin lymphomas harbouring heterogeneous and incompletely delineated molecular aberrations. Over the past decades, molecular studies have identified several chromosomal aberrations, but the interpretation of individual genomic studies can be challenging. OBJECTIVE With a comprehensive meta-analysis, we aim to delineate genomic alterations for different types of CL and propose a more accurate classification in line with their various pathogenicity. METHODS We searched PubMed and ISI Web of Knowledge for publications from 1996 to 2016 reporting the investigation of CL for genome-wide copy number alterations, by means of comparative genomic hybridization techniques and whole-genome sequencing and whole-exome sequencing. We then extracted and remapped the available copy number variation (CNV) data from these publications with the same pipeline and performed clustering and visualisation to aggregate samples of similar CNV profiles. RESULTS For 449 samples from 22 publications, CNV data were accessible for sample based meta-analysis. Our findings illustrate structural and numerical chromosomal imbalance patterns. Most frequent CNAs were linked to oncogenes or tumour suppressor genes with important roles in the course of the disease. CONCLUSION Summary profiles for genomic imbalances, generated from case-specific data, identified complex genomic imbalances, which could discriminate between different subtypes of CL and promise a more accurate classification. The collected data presented in this study are publicly available through the 'Progenetix' online repository.
Collapse
Affiliation(s)
- G Gug
- University of Medicine and Pharmacy "Victor Babeș", Timișoara, România
| | - Q Huang
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Swiss Institute of Bioinformatics, Zurich, Switzerland
| | - E Chiticariu
- University of Medicine and Pharmacy "Victor Babeș", Timișoara, România
| | - C Solovan
- University of Medicine and Pharmacy "Victor Babeș", Timișoara, România.,Emergency City Hospital, University Clinic of Dermatology and Venereology, Timișoara, România
| | - M Baudis
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Swiss Institute of Bioinformatics, Zurich, Switzerland
| |
Collapse
|
26
|
Kohnken R, Mishra A. MicroRNAs in Cutaneous T-Cell Lymphoma: The Future of Therapy. J Invest Dermatol 2019; 139:528-534. [PMID: 30686578 DOI: 10.1016/j.jid.2018.10.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/22/2018] [Accepted: 10/08/2018] [Indexed: 12/20/2022]
Abstract
MicroRNAs (miRs) are small, noncoding RNAs with numerous cellular functions. With advancing knowledge of the many functions of miRs in cancer pathogenesis, there is emerging interest in miRs as therapeutic targets in cancers. One disease that poses an intriguing model for miR therapy is cutaneous T-cell lymphoma, a rare disease featuring malignant CD4+ T cells that proliferate in the skin. The hallmark of cutaneous T-cell lymphoma progression is epigenetic dysregulation, with aberrant miR levels being a common feature. This review aims to summarize the rapidly emerging advances in the development of miR-based therapies in cancers, with a special emphasis on CTCL.
Collapse
Affiliation(s)
- Rebecca Kohnken
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Anjali Mishra
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA; Division of Dermatology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio USA; Department of Medical Oncology, Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
27
|
Abstract
Cutaneous T-cell lymphomas (CTCLs) are a heterogeneous group of lymphomas that are characterized by primary skin involvement. Mycosis fungoides (MF) and Sézary syndrome (SS), the two most common subtypes of CTCL, can be difficult to manage clinically as there are few effective treatment options available. Recently, histone deacetylase inhibitors (HDACi) have emerged as promising therapies with favorable adverse effect profiles, compared with traditional chemotherapies. In this article, we review the published literature to evaluate the role of HDACi in the treatment of CTCL. Specifically, we (1) briefly discuss the molecular rationale for the use of HDACi in CTCL; (2) compare the efficacy, tolerability, and adverse effects of HDACi; (3) review the cardiac safety data; and (4) discuss optimization of therapy with HDACi in the treatment of CTCL.
Collapse
|
28
|
Bastidas Torres AN, Cats D, Mei H, Szuhai K, Willemze R, Vermeer MH, Tensen CP. Genomic analysis reveals recurrent deletion of JAK-STAT signaling inhibitors HNRNPK and SOCS1 in mycosis fungoides. Genes Chromosomes Cancer 2018; 57:653-664. [PMID: 30144205 PMCID: PMC6282857 DOI: 10.1002/gcc.22679] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/21/2018] [Accepted: 08/23/2018] [Indexed: 01/31/2023] Open
Abstract
Mycosis fungoides (MF) is the most common cutaneous T-cell lymphoma (CTCL). Causative genetic alterations in MF are unknown. The low recurrence of pathogenic small-scale mutations (ie, nucleotide substitutions, indels) in the disease, calls for the study of additional aspects of MF genetics. Here, we investigated structural genomic alterations in tumor-stage MF by integrating whole-genome sequencing and RNA-sequencing. Multiple genes with roles in cell physiology (n = 113) and metabolism (n = 92) were found to be impacted by genomic rearrangements, including 47 genes currently implicated in cancer. Fusion transcripts involving genes of interest such as DOT1L, KDM6A, LIFR, TP53, and TP63 were also observed. Additionally, we identified recurrent deletions of genes involved in cell cycle control, chromatin regulation, the JAK-STAT pathway, and the PI-3-K pathway. Remarkably, many of these deletions result from genomic rearrangements. Deletion of tumor suppressors HNRNPK and SOCS1 were the most frequent genetic alterations in MF after deletion of CDKN2A. Notably, SOCS1 deletion could be detected in early-stage MF. In agreement with the observed genomic alterations, transcriptome analysis revealed up-regulation of the cell cycle, JAK-STAT, PI-3-K and developmental pathways. Our results position inactivation of HNRNPK and SOCS1 as potential driver events in MF development.
Collapse
Affiliation(s)
| | - Davy Cats
- Sequencing Analysis Support Core, Leiden University Medical Center, Leiden, The Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, Leiden, The Netherlands
| | - Karoly Szuhai
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rein Willemze
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maarten H Vermeer
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelis P Tensen
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
29
|
Manso R, Martínez-Magunacelaya N, Eraña-Tomás I, Monsalvez V, Rodríguez-Peralto JL, Ortiz-Romero PL, Santonja C, Cristóbal I, Piris MA, Rodríguez-Pinilla SM. Mycosis fungoides progression could be regulated by microRNAs. PLoS One 2018; 13:e0198477. [PMID: 29894486 PMCID: PMC5997347 DOI: 10.1371/journal.pone.0198477] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/18/2018] [Indexed: 12/26/2022] Open
Abstract
Differentiating early mycosis fungoides (MF) from inflammatory dermatitis is a challenge. We compare the differential expression profile of early-stage MF samples and benign inflammatory dermatoses using microRNA (miRNA) arrays. 114 miRNAs were found to be dysregulated between these entities. The seven most differentially expressed miRNAs between these two conditions were further analyzed using RT-PCR in two series comprising 38 samples of early MFs and 18 samples of inflammatory dermatitis. A series of 51 paraffin-embedded samples belonging to paired stages of 16 MF patients was also analyzed. MiRNAs 26a, 222, 181a and 146a were differentially expressed between tumoral and inflammatory conditions. Two of these miRNAs (miRNA-181a and miRNA-146a) were significantly deregulated between early and advanced MF stages. Bioinformatic analysis showed FOXP3 expression to be regulated by these miRNAs. Immunohistochemistry revealed the level of FOXP3 expression to be lower in tumoral MFs than in plaque lesions in paraffin-embedded tissue. A functional study confirmed that both miRNAs diminished FOXP3 expression when overexpressed in CTCL cells. The data presented here suggest that the analysis of a restricted number of miRNAs (26a, 222, 181a and 146a) could be sufficient to differentiate tumoral from reactive conditions. Moreover, these miRNAs seem to be involved in MF progression.
Collapse
Affiliation(s)
- Rebeca Manso
- Pathology Department, Fundación Jiménez Díaz, UAM, Madrid, CIBERONC, Madrid, Spain
| | | | | | - Verónica Monsalvez
- Hospital Universitario 12 de Octubre, Dermatology Department, Madrid, Spain
| | | | | | - Carlos Santonja
- Pathology Department, Fundación Jiménez Díaz, UAM, Madrid, CIBERONC, Madrid, Spain
| | - Ion Cristóbal
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain
| | - Miguel A. Piris
- Pathology Department, Fundación Jiménez Díaz, UAM, Madrid, CIBERONC, Madrid, Spain
| | | |
Collapse
|
30
|
Wilcox RA. Cutaneous T-cell lymphoma: 2017 update on diagnosis, risk-stratification, and management. Am J Hematol 2017; 92:1085-1102. [PMID: 28872191 DOI: 10.1002/ajh.24876] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 07/27/2017] [Indexed: 12/12/2022]
Abstract
DISEASE OVERVIEW Cutaneous T-cell lymphomas are a heterogenous group of T-cell lymphoproliferative disorders involving the skin, the majority of which may be classified as Mycosis Fungoides (MF) or Sézary Syndrome (SS). DIAGNOSIS The diagnosis of MF or SS requires the integration of clinical and histopathologic data. RISK-ADAPTED THERAPY TNMB (tumor, node, metastasis, blood) staging remains the most important prognostic factor in MF/SS and forms the basis for a "risk-adapted," multi-disciplinary approach to treatment. For patients with disease limited to the skin, expectant management or skin-directed therapies is preferred, as both disease-specific and overall survival for these patients is favorable. In contrast, patients with advanced-stage disease with significant nodal, visceral or blood involvement are generally approached with biologic-response modifiers or histone deacetylase inhibitors prior to escalating therapy to include systemic, single-agent chemotherapy. In highly-selected patients, allogeneic stem-cell transplantation may be considered, as this may be curative in some patients.
Collapse
Affiliation(s)
- Ryan A. Wilcox
- Division of Hematology/Oncology; University of Michigan Comprehensive Cancer Center; Ann Arbor Michigan 48109-5948
| |
Collapse
|
31
|
Ni X, Zhang X, Hu CH, Langridge T, Tarapore RS, Allen JE, Oster W, Duvic M. ONC201 selectively induces apoptosis in cutaneous T-cell lymphoma cells via activating pro-apoptotic integrated stress response and inactivating JAK/STAT and NF-κB pathways. Oncotarget 2017; 8:61761-61776. [PMID: 28977902 PMCID: PMC5617462 DOI: 10.18632/oncotarget.18688] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 05/22/2017] [Indexed: 11/25/2022] Open
Abstract
Cutaneous T-cell lymphomas (CTCLs) are extremely symptomatic and still incurable, and more effective and less toxic therapies are urgently needed. ONC201, an imipridone compound, has shown efficacy in pre-clinical studies in multiple advanced cancers. This study was to evaluate the anti-tumor activity of ONC201 on CTCL cells. The effect of ONC201 on the cell growth and apoptosis were evaluated in CTCL cell lines (n=8) and primary CD4+ malignant T cells isolated from CTCL patients (n=5). ONC201 showed a time-dependent cell growth inhibition in all treated cell lines with a concentration range of 1.25-10.0 μM. ONC201 also induced apoptosis in tested cells with a narrow concentration range of 2.5-10.0 μM, evidenced by increased Annexin V+ cells, accompanied by accumulated sub-G1 portions. ONC201 only induced apoptosis in CD4+ malignant T cells, not in normal CD4+ T cells. The activating transcription factor 4 (ATF4), a hallmark of integrated stress response, was upregulated in response to ONC201 whereas Akt was downregulated. In addition, molecules in JAK/STAT and NF-κB pathways, as well as IL-32β, were downregulated following ONC201 treatment. Thus, ONC201 exerts a potent and selective anti-tumor effect on CTCL cells. Its efficacy may involve activating integrated stress response through ATF4 and inactivating JAK/STAT and NF-κB pathways.
Collapse
Affiliation(s)
- Xiao Ni
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiang Zhang
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cheng-Hui Hu
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy Langridge
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Madeleine Duvic
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
32
|
Bowman T, Yu H, Sebti S, Dalton W, Jove R. Signal Transducers and Activators of Transcription: Novel Targets for Anticancer Therapeutics. Cancer Control 2017. [DOI: 10.1177/107327489900600501] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Through specific activation of gene expression, the family of proteins known as signal transducers and activators of transcription (STATs) converts extracellular stimuli into diverse biological responses. Beyond the normal signaling functions of STATs, recent evidence indicates that aberrant activation of STATs contributes to neoplastic transformation. Methods Current literature pertaining to the role of STAT proteins in oncogenesis is presented. Also, the rationale for developing novel approaches to disrupt STAT signaling is discussed, and the potential of STATs as anticancer targets in treating human cancer is reviewed. Results The discovery that certain oncoproteins constitutively activate specific STATs, coupled with observations that elevated STAT activity occurs frequently in a spectrum of human tumors, establishes a direct link between STAT activation and neoplastic transformation. Significantly, abrogation of STAT signaling blocks oncogenesis in model in vitro and in vivo systems. These results make STATs attractive targets for rational design of small molecule inhibitors and gene therapy approaches to disrupt STAT signaling. Conclusions As a result of genetic, biochemical, and crystallographic analyses, the functional domains of STAT proteins have been well characterized. Based on these data, selective inhibitors of STAT function can be designed. Because disrupting STAT signaling has proven effective in blocking neoplastic transformation, it is proposed that STAT proteins represent promising targets for development of novel molecular therapeutics to treat human cancer.
Collapse
Affiliation(s)
- Tammy Bowman
- Molecular Oncology Program at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Fla
| | - Hua Yu
- Immunology Program at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Fla
| | - Saïd Sebti
- Drug Discovery Program at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Fla
| | - William Dalton
- Clinical Investigations Program at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Fla
| | - Richard Jove
- Molecular Oncology Program at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Fla
| |
Collapse
|
33
|
Ma C, Horlad H, Pan C, Yano H, Ohnishi K, Fujiwara Y, Matsuoka M, Lee A, Niidome T, Yamanaka R, Takeya M, Komohara Y. Stat3 inhibitor abrogates the expression of PD-1 ligands on lymphoma cell lines. J Clin Exp Hematop 2017; 57:21-25. [PMID: 28496056 DOI: 10.3960/jslrt.17006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Recent studies have indicated the significance of immune checkpoint molecules including programmed death-1 (PD-1), cytotoxic T-lymphocyte associated protein 4, and T-cell immunoglobulin and mucin domain-containing molecule-3 for anti-tumor immune responses. We previously investigated PD-1 ligand 1/2 (PD-L1/2) expression in lymphoma cell lines, and found that PD-L1/2 is expressed on the adult T-cell leukemia/lymphoma (ATL-T) and B-cell lymphoma (SLVL) cell lines. In the present study, we investigated whether the Stat3 inhibitor WP1066 abrogated PD-L1/2 expression in lymphoma cell lines. Incubation with WP1066 inhibited lymphoma cell growth and induced cell apoptosis. PD-L1/2 expression in the ATL-T, SLVL, and human brain malignant lymphoma (HKBML) cell lines was significantly abrogated by WP1066 treatment. These data indicated that a Stat3 inhibitor abrogated PD-L1/2 expression in lymphoma cells. Such an inhibitor is therefore considered to be useful for additional immunotherapy in patients with advanced lymphoma.
Collapse
Affiliation(s)
- Chaoya Ma
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Hasita Horlad
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Cheng Pan
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Hiromu Yano
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Koji Ohnishi
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Masao Matsuoka
- Department of Hematology, Graduate School of Medical Sciences, Kumamoto University.,Laboratory of Virus Control, Institute for Virus Research, Kyoto University
| | - Aeju Lee
- International Research Organization for Advanced Science and Technology (IROAST), Kumamoto University.,Magnesium Research Center, Kumamoto University
| | - Takuro Niidome
- Faculty of Advanced Science and Technology, Kumamoto University
| | - Ryuya Yamanaka
- Kyoto Prefectural University of Medicine, Graduate School for Health Care Science
| | - Motohiro Takeya
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University
| |
Collapse
|
34
|
Mahony R, Gargan S, Roberts KL, Bourke N, Keating SE, Bowie AG, O'Farrelly C, Stevenson NJ. A novel anti-viral role for STAT3 in IFN-α signalling responses. Cell Mol Life Sci 2017; 74:1755-1764. [PMID: 27988795 PMCID: PMC11107673 DOI: 10.1007/s00018-016-2435-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 11/16/2016] [Accepted: 12/05/2016] [Indexed: 10/20/2022]
Abstract
The cytokine, Interferon (IFN)-α, induces a wide spectrum of anti-viral mediators, via the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway. STAT1 and STAT2 are well characterised to upregulate IFN-stimulated gene (ISG) expression; but even though STAT3 is also activated by IFN-α, its role in anti-viral ISG induction is unclear. Several viruses, including Hepatitis C and Mumps, reduce cellular STAT3 protein levels, via the promotion of ubiquitin-mediated proteasomal degradation. This viral immune evasion mechanism suggests an undiscovered anti-viral role for STAT3 in IFN-α signalling. To investigate STAT3's functional involvement in this Type I IFN pathway, we first analysed its effect upon the replication of two viruses, Influenza and Vaccinia. Viral plaque assays, using Wild Type (WT) and STAT3-/- Murine Embryonic Fibroblasts (MEFs), revealed that STAT3 is required for the inhibition of Influenza and Vaccinia replication. Furthermore, STAT3 shRNA knockdown also enhanced Influenza replication and hindered induction of several, well characterised, anti-viral ISGs: PKR, OAS2, MxB and ISG15; while STAT3 expression had no effect upon induction of a separate ISG group: Viperin, IFI27, CXCL10 and CCL5. These discoveries reveal, for the first time, an anti-viral role for STAT3 in the IFN-α pathway and characterise a requirement for STAT3 in the expression of specific ISGs. These findings also identify STAT3 as a therapeutic target against viral infection and highlight it as an essential pathway component for endogenous and therapeutic IFN-α responsiveness.
Collapse
Affiliation(s)
- Rebecca Mahony
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Siobhán Gargan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Kim L Roberts
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Nollaig Bourke
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Sinead E Keating
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
| | - Nigel J Stevenson
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
35
|
Shreberk-Hassidim R, Ramot Y, Zlotogorski A. Janus kinase inhibitors in dermatology: A systematic review. J Am Acad Dermatol 2017; 76:745-753.e19. [DOI: 10.1016/j.jaad.2016.12.004] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/29/2016] [Accepted: 12/06/2016] [Indexed: 02/08/2023]
|
36
|
STAT3 Controls the Long-Term Survival and Phenotype of Repair Schwann Cells during Nerve Regeneration. J Neurosci 2017; 37:4255-4269. [PMID: 28320842 PMCID: PMC5413174 DOI: 10.1523/jneurosci.3481-16.2017] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/11/2017] [Accepted: 01/19/2017] [Indexed: 11/21/2022] Open
Abstract
After nerve injury, Schwann cells convert to a phenotype specialized to promote repair. But during the slow process of axonal regrowth, these repair Schwann cells gradually lose their regeneration-supportive features and eventually die. Although this is a key reason for the frequent regeneration failures in humans, the transcriptional mechanisms that control long-term survival and phenotype of repair cells have not been studied, and the molecular signaling underlying their decline is obscure. We show, in mice, that Schwann cell STAT3 has a dual role. It supports the long-term survival of repair Schwann cells and is required for the maintenance of repair Schwann cell properties. In contrast, STAT3 is less important for the initial generation of repair Schwann cells after injury. In repair Schwann cells, we find that Schwann cell STAT3 activation by Tyr705 phosphorylation is sustained during long-term denervation. STAT3 is required for maintaining autocrine Schwann cell survival signaling, and inactivation of Schwann cell STAT3 results in a striking loss of repair cells from chronically denervated distal stumps. STAT3 inactivation also results in abnormal morphology of repair cells and regeneration tracks, and failure to sustain expression of repair cell markers, including Shh, GDNF, and BDNF. Because Schwann cell development proceeds normally without STAT3, the function of this factor appears restricted to Schwann cells after injury. This identification of transcriptional mechanisms that support long-term survival and differentiation of repair cells will help identify, and eventually correct, the failures that lead to the deterioration of this important cell population. SIGNIFICANCE STATEMENT Although injured peripheral nerves contain repair Schwann cells that provide signals and spatial clues for promoting regeneration, the clinical outcome after nerve damage is frequently poor. A key reason for this is that, during the slow growth of axons through the proximal parts of injured nerves repair, Schwann cells gradually lose regeneration-supporting features and eventually die. Identification of signals that sustain repair cells is therefore an important goal. We have found that in mice the transcription factor STAT3 protects these cells from death and contributes to maintaining the molecular and morphological repair phenotype that promotes axonal regeneration. Defining the molecular mechanisms that maintain repair Schwann cells is an essential step toward developing therapeutic strategies that improve nerve regeneration and functional recovery.
Collapse
|
37
|
Ferraz ERA, Fernandes AS, Salviano I, Felzenszwalb I, Mencalha AL. Investigation of the mutagenic and genotoxic activities of LLL-3, a STAT3 inhibitor. Drug Chem Toxicol 2017; 40:30-35. [PMID: 28140701 DOI: 10.3109/01480545.2016.1167901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
LLL-3, an anthracene derived compound, has been shown to be a promising therapeutic agent for the treatment of some kinds of cancer such as chronic myeloid leukemia and glioblastoma. However, no data regarding the toxic properties of this compound have yet been described in the literature. The present work aimed to investigate the mutagenic and genotoxic activities of LLL-3 using the TA97, TA98, TA100, TA102 and TA104 Salmonella/microsome strains for the Ames test and the micronucleus assay with the mouse macrophage cell line RAW 264.7. The findings showed that LLL-3, at doses of 0.001, 0.01, 0.1, 1.0 and 10.0 μg/plate, did not induce mutagenic activity in the Salmonella strains used under the conditions tested, and nor did it present genotoxicity in RAW 264.7 cells, at 10.0, 100.0 and 1000.0 μg/mL doses. Moreover, it is important to point out that the mitotic index of the cells decreased after exposure to LLL-3 under the same conditions tested, which may suggest some cytostatic effect, since this compound acts by inhibiting STAT3. Since most drugs used in the treatment of cancer present mutagenic activity as an adverse effect, these results suggest that LLL-3 is a promising drug for cancer therapy.
Collapse
Affiliation(s)
- E R A Ferraz
- a Environmental Mutagenesis Laboratory, Department of Biophysics and Biometry, Roberto Alcantra Gomes Biology Institute, University of the State of Rio de Janeiro , Rio de Janeiro , RJ , Brazil.,b School of Pharmacy, Fluminense Federal University , Niteroi , RJ , Brazil , and
| | - A S Fernandes
- a Environmental Mutagenesis Laboratory, Department of Biophysics and Biometry, Roberto Alcantra Gomes Biology Institute, University of the State of Rio de Janeiro , Rio de Janeiro , RJ , Brazil
| | - I Salviano
- c Laboratory of Cancer Biology , Department of Biophysics and Biometry, Roberto Alcantra Gomes Biology Institute, University of the State of Rio de Janeiro , Rio de Janeiro , RJ , Brazil
| | - I Felzenszwalb
- a Environmental Mutagenesis Laboratory, Department of Biophysics and Biometry, Roberto Alcantra Gomes Biology Institute, University of the State of Rio de Janeiro , Rio de Janeiro , RJ , Brazil
| | - A L Mencalha
- c Laboratory of Cancer Biology , Department of Biophysics and Biometry, Roberto Alcantra Gomes Biology Institute, University of the State of Rio de Janeiro , Rio de Janeiro , RJ , Brazil
| |
Collapse
|
38
|
Chen Q, Gu Y, Zhang S, Deng H. Effects and mechanisms of action of SARI on androgen-independent prostate cancer (DU145) cells. Tumour Biol 2016; 37:16141–16149. [PMID: 27739031 DOI: 10.1007/s13277-016-5469-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/23/2016] [Indexed: 12/14/2022] Open
Abstract
This study aimed to characterize the role and mechanisms of action of suppressor of AP-1, regulated by IFN (SARI) in androgen-independent prostate cancer cells using the DU145 cell line. Prostate cancer cell lines were transfected to permit both the overexpression and inhibition of SARI. MTT assays and Transwell assays were performed to detect the effects of SARI overexpression and inhibition on the proliferation activity, invasiveness, and metastatic ability of DU145 cells. Expression of vascular endothelial growth factor (VEGF) and tyrosine-phosphorylated signal transducer and activator of transcription 3 (p-STAT3) was monitored in the experimental groups using a qPCR assay and western blot analysis. Additionally, DU145 cells were separately treated with 5, 50, and 100 μmol/L AG490 for 48 h and SARI expression was detected using the qPCR assay and western blot analysis. We also monitored the effects of AG490 treatment (100 μmol/L for 48 h) on both the SARI-SiRNA DU145 cells and empty vector DU145 (DU145-V) cells using the MTT assay and a Transwell migration assay. SARI overexpression and SARI-SiRNA DU145 prostate cancer cell lines were successfully established. The proliferation activity and the invasion and migration abilities of DU145-SARI cells were significantly lower compared with the DU145-V group (P < 0.05). Conversely, the proliferation activity and the invasion and migration abilities of SARI-SiRNA cells were significantly higher compared with the DU145-V group (P < 0.05). VEGF and p-STAT3 expression levels were lower in the SARI overexpression group compared with the DU145-V group and the control group (P < 0.05). In contrast, VEGF and p-STAT3 expression levels were higher in the SARI-SiRNA group compared with both the DU145-V group and the control group (P < 0.05). In comparison with the control group, SARI expression levels were higher in DU145 cells treated with 50 and 100 μmol/L AG490. Upon treatment with 100 μmol/L AG490 for 48 h, the proliferation activity and invasiveness and migration abilities of SARI-SiRNA cells were significantly higher compared with the DU145-V group (P < 0.05). SARI significantly affects the proliferation, invasion, and metastasis of DU145 cells. It is possible that SARI inhibits the proliferation, invasion, and migration of androgen-independent prostate cancer cells by regulating downstream genes through the SARI/STAT3/VEGF pathways.
Collapse
Affiliation(s)
- Qian Chen
- Department of Pathology, The Third Xiangya Hospital, The Central South University, Changsha, Hunan, China
| | - Yonghong Gu
- Department of Pathology, The Third Xiangya Hospital, The Central South University, Changsha, Hunan, China.
| | - Shengwang Zhang
- Department of radiology, The Third Xiangya Hospital, The Central South University, Changsha, Hunan, China
| | - Hao Deng
- Center for Experimental Medicine, The Third Xiangya Hospital, The Central South University, Changsha, Hunan, China
| |
Collapse
|
39
|
Ma H, Abdul-Hay M. T-cell lymphomas, a challenging disease: types, treatments, and future. Int J Clin Oncol 2016; 22:18-51. [PMID: 27743148 PMCID: PMC7102240 DOI: 10.1007/s10147-016-1045-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/26/2016] [Indexed: 02/06/2023]
Abstract
T-cell lymphomas are rare and aggressive malignancies associated with poor outcome, often because of the development of resistance in the lymphoma against chemotherapy as well as intolerance in patients to the established and toxic chemotherapy regimens. In this review article, we discuss the epidemiology, pathophysiology, current standard of care, and future treatments of common types of T-cell lymphomas, including adult T-cell leukemia/lymphoma, angioimmunoblastic T-cell lymphoma, anaplastic large-cell lymphoma, aggressive NK/T-cell lymphoma, and cutaneous T-cell lymphoma.
Collapse
Affiliation(s)
- Helen Ma
- Department of Internal Medicine, New York University, New York, NY, USA
| | - Maher Abdul-Hay
- Department of Internal Medicine, New York University, New York, NY, USA. .,Perlmutter Cancer Center, New York University, New York, NY, USA.
| |
Collapse
|
40
|
Dong W, Xian Y, Yuan W, Huifeng Z, Tao W, Zhiqiang L, Shan F, Ya F, Hongli W, Jinghuan W, Lei Q, Li Z, Hongyi Q. Catalpol stimulates VEGF production via the JAK2/STAT3 pathway to improve angiogenesis in rats' stroke model. JOURNAL OF ETHNOPHARMACOLOGY 2016; 191:169-179. [PMID: 27301615 DOI: 10.1016/j.jep.2016.06.030] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/05/2016] [Accepted: 06/08/2016] [Indexed: 05/25/2023]
Abstract
ETHNOBOTANICAL RELEVANCE Catalpol is the main active component of the radix from Rehmannia glutinosa Libosch, which has pleiotropic protective effects in neurodegenerative diseases, ischemic stroke, metabolic disorders and others AIM Catalpol has been shown to have neuroprotective, neurorepair, and angiogenesis effects following ischemic brain injury. However, its molecular mechanisms are still poorly understood. In previous studies, the JAK2/STAT3 signaling pathway was found to play a role in neuroprotection and angiogenesis. This study investigated the role of catalpol in stimulating angiogenesis via the JAK2/STAT3 pathway after permanent focal cerebral ischemia (pMCAO). METHODS Rats were subjected to right middle cerebral artery occlusion through electrocoagulation and were treated with catalpol (5mg/kg), AG490 was also used to inhibit STAT3 phosphorylation (pSTAT3). RESULTS Following stroke, Catalpol improved the neuroethology deficit, increased the cerebral blood flow (CBF) of infarcted brain and upregulated EPO and EPOR. AG490 suppressed the phosphorylation of signal transducer and activator of transcription 3 (STAT3), ultimately inhibited VEGF mRNA expression, which reduced VEGF protein expression and inhibited stroke-induced angiogenesis. However, Catalpol enhanced stroke-induced STAT3 activation and subsequently restored STAT3 activity through the recovery of STAT3 binding to VEGF. Moreover, Catalpol reversed the effect of AG490 on STAT3 activation and nuclear translocation, restored the transcriptional activity of the VEGF promoter by recruiting STAT3 to the VEGF promoter, improved VEGF mRNA and protein expression, increased angiogenesis, reduced the difference in CBF between the infarcted and intact brain and ameliorated the neuroethology behaviors after stroke. CONCLUSION Catalpol affects neuroprotection and angiogenesis via the JAK2/STAT3 signaling pathway, which is mediated by STAT3 activation and VEGF expression. Catalpol may be used as a potential therapeutic drug for stroke.
Collapse
MESH Headings
- Angiogenesis Inducing Agents/pharmacology
- Animals
- Brain/drug effects
- Brain/enzymology
- Brain/pathology
- Brain/physiopathology
- Cerebral Arteries/drug effects
- Cerebral Arteries/enzymology
- Cerebral Arteries/pathology
- Cerebral Arteries/physiopathology
- Cerebrovascular Circulation/drug effects
- Disease Models, Animal
- Erythropoietin/metabolism
- Infarction, Middle Cerebral Artery/drug therapy
- Infarction, Middle Cerebral Artery/enzymology
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/physiopathology
- Iridoid Glucosides/pharmacology
- Janus Kinase 2/metabolism
- Male
- Neovascularization, Physiologic/drug effects
- Neuroprotective Agents/pharmacology
- Phosphorylation
- Promoter Regions, Genetic
- Protein Binding
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptors, Erythropoietin/metabolism
- STAT3 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Time Factors
- Transcriptional Activation
- Up-Regulation
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Wan Dong
- Department of Emergency, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yang Xian
- Department of Pharmacy, The Seventh People's Hospital of Chengdu, Chengdu 610041, China
| | - Wang Yuan
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Zhu Huifeng
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China.
| | - Wang Tao
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Liu Zhiqiang
- Department of Pharmacy, The First People's Hospital of Neijiang, Neijiang 641000, China
| | - Feng Shan
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Fu Ya
- College of Chemistry and Chemical Engineering, Chongqing University of Science & Technology, Chongqing 401331, China
| | - Wang Hongli
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Wang Jinghuan
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Qin Lei
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Zou Li
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Qi Hongyi
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China; Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| |
Collapse
|
41
|
Sibbesen NA, Kopp KL, Litvinov IV, Jønson L, Willerslev-Olsen A, Fredholm S, Petersen DL, Nastasi C, Krejsgaard T, Lindahl LM, Gniadecki R, Mongan NP, Sasseville D, Wasik MA, Iversen L, Bonefeld CM, Geisler C, Woetmann A, Odum N. Jak3, STAT3, and STAT5 inhibit expression of miR-22, a novel tumor suppressor microRNA, in cutaneous T-Cell lymphoma. Oncotarget 2016; 6:20555-69. [PMID: 26244872 PMCID: PMC4653025 DOI: 10.18632/oncotarget.4111] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/22/2015] [Indexed: 01/08/2023] Open
Abstract
Aberrant activation of Janus kinase-3 (Jak3) and its key down-stream effectors, Signal Transducer and Activator of Transcription-3 (STAT3) and STAT5, is a key feature of malignant transformation in cutaneous T-cell lymphoma (CTCL). However, it remains only partially understood how Jak3/STAT activation promotes lymphomagenesis. Recently, non-coding microRNAs (miRNAs) have been implicated in the pathogenesis of this malignancy. Here, we show that (i) malignant T cells display a decreased expression of a tumor suppressor miRNA, miR-22, when compared to non-malignant T cells, (ii) STAT5 binds the promoter of the miR-22 host gene, and (iii) inhibition of Jak3, STAT3, and STAT5 triggers increased expression of pri-miR-22 and miR-22. Curcumin, a nutrient with anti-Jak3 activity and histone deacetylase inhibitors (HDACi) also trigger increased expression of pri-miR-22 and miR-22. Transfection of malignant T cells with recombinant miR-22 inhibits the expression of validated miR-22 targets including NCoA1, a transcriptional co-activator in others cancers, as well as HDAC6, MAX, MYCBP, PTEN, and CDK2, which have all been implicated in CTCL pathogenesis. In conclusion, we provide the first evidence that de-regulated Jak3/STAT3/STAT5 signalling in CTCL cells represses the expression of the gene encoding miR-22, a novel tumor suppressor miRNA.
Collapse
Affiliation(s)
- Nina A Sibbesen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Katharina L Kopp
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Ivan V Litvinov
- Division of Dermatology, McGill University Health Centre, Montréal, Quebec, Canada
| | - Lars Jønson
- Departmen of Molecular Medicine, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | | | - Simon Fredholm
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - David L Petersen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Claudia Nastasi
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Thorbjørn Krejsgaard
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Lise M Lindahl
- Department of Dermatology, Aarhus University Hospital, Skejby, Aarhus, Denmark
| | - Robert Gniadecki
- Departmen of Dermatology, Copenhagen University Hospital, Bispebjerg, Copenhagen, Denmark
| | - Nigel P Mongan
- Faculty of Medicine and Health Science, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Denis Sasseville
- Division of Dermatology, McGill University Health Centre, Montréal, Quebec, Canada
| | - Mariusz A Wasik
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lars Iversen
- Department of Dermatology, Aarhus University Hospital, Skejby, Aarhus, Denmark
| | - Charlotte M Bonefeld
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Geisler
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Anders Woetmann
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Niels Odum
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
42
|
Staphylococcal enterotoxin A (SEA) stimulates STAT3 activation and IL-17 expression in cutaneous T-cell lymphoma. Blood 2016; 127:1287-96. [PMID: 26738536 DOI: 10.1182/blood-2015-08-662353] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 12/01/2015] [Indexed: 12/23/2022] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) is characterized by proliferation of malignant T cells in a chronic inflammatory environment. With disease progression, bacteria colonize the compromised skin barrier and half of CTCL patients die of infection rather than from direct organ involvement by the malignancy. Clinical data indicate that bacteria play a direct role in disease progression, but little is known about the mechanisms involved. Here, we demonstrate that bacterial isolates containing staphylococcal enterotoxin A (SEA) from the affected skin of CTCL patients, as well as recombinant SEA, stimulate activation of signal transducer and activator of transcription 3 (STAT3) and upregulation of interleukin (IL)-17 in immortalized and primary patient-derived malignant and nonmalignant T cells. Importantly, SEA induces STAT3 activation and IL-17 expression in malignant T cells when cocultured with nonmalignant T cells, indicating an indirect mode of action. In accordance, malignant T cells expressing an SEA-nonresponsive T-cell receptor variable region β chain are nonresponsive to SEA in monoculture but display strong STAT3 activation and IL-17 expression in cocultures with SEA-responsive nonmalignant T cells. The response is induced via IL-2 receptor common γ chain cytokines and a Janus kinase 3 (JAK3)-dependent pathway in malignant T cells, and blocked by tofacitinib, a clinical-grade JAK3 inhibitor. In conclusion, we demonstrate that SEA induces cell cross talk-dependent activation of STAT3 and expression of IL-17 in malignant T cells, suggesting a mechanism whereby SEA-producing bacteria promote activation of an established oncogenic pathway previously implicated in carcinogenesis.
Collapse
|
43
|
Wilcox RA. Cutaneous T-cell lymphoma: 2016 update on diagnosis, risk-stratification, and management. Am J Hematol 2016; 91:151-65. [PMID: 26607183 PMCID: PMC4715621 DOI: 10.1002/ajh.24233] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 11/03/2015] [Indexed: 12/11/2022]
Abstract
DISEASE OVERVIEW Cutaneous T-cell lymphomas are a heterogenous group of T-cell lymphoproliferative disorders involving the skin, the majority of which may be classified as Mycosis Fungoides (MF) or Sézary Syndrome (SS). DIAGNOSIS The diagnosis of MF or SS requires the integration of clinical and histopathologic data. RISK-ADAPTED THERAPY TNMB (tumor, node, metastasis, blood) staging remains the most important prognostic factor in MF/SS and forms the basis for a "risk-adapted," multidisciplinary approach to treatment. For patients with disease limited to the skin, expectant management or skin-directed therapies is preferred, as both disease-specific and overall survival for these patients is favorable. In contrast, patients with advanced-stage disease with significant nodal, visceral, or blood involvement are generally approached with biologic-response modifiers or histone deacetylase inhibitors before escalating therapy to include systemic, single-agent chemotherapy. In highly-selected patients, allogeneic stem-cell transplantation may be considered, as this may be curative in some patients.
Collapse
Affiliation(s)
- Ryan A. Wilcox
- Division of Hematology/Oncology, University of Michigan Cancer Center, 1500 E. Medical Center Drive, Room 4310 CC, Ann Arbor, MI 48109-5948
| |
Collapse
|
44
|
Yamauchi A, Itaya-Hironaka A, Sakuramoto-Tsuchida S, Takeda M, Yoshimoto K, Miyaoka T, Fujimura T, Tsujinaka H, Tsuchida C, Ota H, Takasawa S. Synergistic activations of REG I α and REG I β promoters by IL-6 and Glucocorticoids through JAK/STAT pathway in human pancreatic β cells. J Diabetes Res 2015; 2015:173058. [PMID: 25767811 PMCID: PMC4342170 DOI: 10.1155/2015/173058] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/26/2015] [Indexed: 12/31/2022] Open
Abstract
Reg (Regenerating gene) gene was originally isolated from rat regenerating islets and its encoding protein was revealed as an autocrine/paracrine growth factor for β cells. Rat Reg gene is activated in inflammatory conditions for β cell regeneration. In human, although five functional REG family genes (REG Iα, REG Iβ, REG III, HIP/PAP, and REG IV) were isolated, their expressions in β cells under inflammatory conditions remained unclear. In this study, we found that combined addition of IL-6 and dexamethasone (Dx) induced REG Iα and REG Iβ expression in human 1.1B4 β cells. Promoter assay revealed that a signal transducer and activator of transcription- (STAT-) binding site in each promoter of REG Iα (TGCCGGGAA) and REG Iβ (TGCCAGGAA) was essential for the IL-6+Dx-induced promoter activation. A Janus kinase 2 (JAK2) inhibitor significantly inhibited the IL-6+Dx-induced REG Iα and REG Iβ transcription. Electrophoretic mobility shift assay and chromatin immunoprecipitation revealed that IL-6+Dx stimulation increased STAT3 binding to the REG Iα promoter. Furthermore, small interfering RNA-mediated targeting of STAT3 blocked the IL-6+Dx-induced expression of REG Iα and REG Iβ. These results indicate that the expression of REG Iα and REG Iβ should be upregulated in human β cells under inflammatory conditions through the JAK/STAT pathway.
Collapse
Affiliation(s)
- Akiyo Yamauchi
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | | | | | - Maiko Takeda
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Kiyomi Yoshimoto
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Tomoko Miyaoka
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Takanori Fujimura
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Hiroki Tsujinaka
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Chikatsugu Tsuchida
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Hiroyo Ota
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Shin Takasawa
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| |
Collapse
|
45
|
Manku G, Wang Y, Merkbaoui V, Boisvert A, Ye X, Blonder J, Culty M. Role of retinoic acid and platelet-derived growth factor receptor cross talk in the regulation of neonatal gonocyte and embryonal carcinoma cell differentiation. Endocrinology 2015; 156:346-59. [PMID: 25380237 PMCID: PMC5393322 DOI: 10.1210/en.2014-1524] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neonatal gonocytes are direct precursors of spermatogonial stem cells, the cell pool that supports spermatogenesis. Although unipotent in vivo, gonocytes express pluripotency genes common with embryonic stem cells. Previously, we found that all-trans retinoic acid (RA) induced the expression of differentiation markers and a truncated form of platelet-derived growth factor receptor (PDGFR)β in rat gonocytes, as well as in F9 mouse embryonal carcinoma cells, an embryonic stem cell-surrogate that expresses somatic lineage markers in response to RA. The present study is focused on identifying the signaling pathways involved in RA-induced gonocyte and F9 cell differentiation. Mitogen-activated protein kinase kinase (MEK) 1/2 activation was required during F9 cell differentiation towards somatic lineage, whereas its inhibition potentiated RA-induced Stra8 expression, suggesting that MEK1/2 acts as a lineage specification switch in F9 cells. In both cell types, RA increased the expression of the spermatogonial/premeiotic marker Stra8, which is in line with F9 cells being at a stage before somatic-germline lineage specification. Inhibiting PDGFR kinase activity reduced RA-induced Stra8 expression. Interestingly, RA increased the expression of PDGFRα variant forms in both cell types. Together, these results suggest a potential cross talk between RA and PDGFR signaling pathways in cell differentiation. RA receptor-α inhibition partially reduced RA effects on Stra8 in gonocytes, indicating that RA acts in part via RA receptor-α. RA-induced gonocyte differentiation was significantly reduced by inhibiting SRC (v-src avian sarcoma [Schmidt-Ruppin A-2] viral oncogene) and JAK2/STAT5 (Janus kinase 2/signal transducer and activator of transcription 5) activities, implying that these signaling molecules play a role in gonocyte differentiation. These results suggest that gonocyte and F9 cell differentiation is regulated via cross talk between RA and PDGFRs using different downstream pathways.
Collapse
Affiliation(s)
- Gurpreet Manku
- The Research Institute of the McGill University Health Centre (G.M., V.M., A.B., M.C.), Montreal, Quebec, Canada H3G1A4; Departments of Pharmacology and Therapeutics (G.M., M.C.) Medicine (M.C.), McGill University, Montreal, Quebec, Canada H3G1A4; Department of Biochemistry and Molecular and Cellular Biology (Y.W.), Georgetown University Medical Center, Washington, DC 20057; and Protein Characterization Laboratory (X.Y., J.B.), Cancer Research Technology Program, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, Maryland 21702
| | | | | | | | | | | | | |
Collapse
|
46
|
Lu Z, Wang J, Zheng T, Liang Y, Yin D, Song R, Pei T, Pan S, Jiang H, Liu L. FTY720 inhibits proliferation and epithelial-mesenchymal transition in cholangiocarcinoma by inactivating STAT3 signaling. BMC Cancer 2014; 14:783. [PMID: 25344679 PMCID: PMC4221672 DOI: 10.1186/1471-2407-14-783] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 10/17/2014] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Interleukin 6 (IL-6)-mediated signal transducers and activators of transcription 3 (STAT-3) phosphorylation (activation) is aberrantly sustained in cholangiocarcinoma cells resulting in enhanced myeloid cell leukemia 1 (Mcl-1) expression and resistance to apoptosis. FTY720, a new immunosuppressant, derived from ISP-1, has been studied for its putative anti-cancer properties. This study aimed to elucidate the mechanism by which FTY720 mediates antitumor effects in cholangiocarcinoma (CC) cells. METHODS Three CC cell lines were examined, QBC939, TFK-1, and HuCCT1. The therapeutic effects of FTY720 were evaluated in vitro and in vivo. Cell proliferation, apoptosis, cell cycle, invasive potential, and epithelial- mesenchy-mal transition (EMT) were examined. RESULTS FTY720 greatly inhibited CC cells proliferation and EMT in vitro and in vivo, and this effect was associated with dephosphorylation of STAT3tyr705. FTY720 induced apoptosis and G1 phase arrest in CC cells, and inhibited invasion of CC cells. Western blot analysis showed that FTY720 induced cleavage of caspases 3, 8 and 9, and of PARP, in a dose-dependent manner, consistent with a substantial decrease in p-STAT3, Bcl-xL, Bcl-2, survivin, cyclin D1, cyclin E, N-cadherin, vimentin, VEGF and TWIST1. In vivo studies showed that tumor growth and metastasis were significantly suppressed after FTY720 treatment. CONCLUSIONS These results suggest that FTY720 induces a significant decrease in p-STAT3, which inhibits proliferation and EMT of CC cells, and then induces G1 phase arrest and apoptosis. We have characterized a novel immunosuppressant, which shows potential anti-tumor effects on CC via p-STAT3 inhibition. FTY720 merits further investigation and warrants clinical evaluation.
Collapse
Affiliation(s)
- Zhaoyang Lu
- />Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, No. 23 Youzheng Street, Harbin, Heilongjiang Province 150001 China
| | - Jiabei Wang
- />Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, No. 23 Youzheng Street, Harbin, Heilongjiang Province 150001 China
| | - Tongsen Zheng
- />Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, No. 23 Youzheng Street, Harbin, Heilongjiang Province 150001 China
| | - Yingjian Liang
- />Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, No. 23 Youzheng Street, Harbin, Heilongjiang Province 150001 China
| | - Dalong Yin
- />Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, No. 23 Youzheng Street, Harbin, Heilongjiang Province 150001 China
| | - Ruipeng Song
- />Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, No. 23 Youzheng Street, Harbin, Heilongjiang Province 150001 China
| | - Tiemin Pei
- />Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, No. 23 Youzheng Street, Harbin, Heilongjiang Province 150001 China
| | - Shangha Pan
- />Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, No. 23 Youzheng Street, Harbin, Heilongjiang Province 150001 China
| | - Hongchi Jiang
- />Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, No. 23 Youzheng Street, Harbin, Heilongjiang Province 150001 China
| | - Lianxin Liu
- />Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, No. 23 Youzheng Street, Harbin, Heilongjiang Province 150001 China
- />Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, China
| |
Collapse
|
47
|
Kigawa Y, Miyazaki T, Lei XF, Nakamachi T, Oguchi T, Kim-Kaneyama JR, Taniyama M, Tsunawaki S, Shioda S, Miyazaki A. NADPH oxidase deficiency exacerbates angiotensin II-induced abdominal aortic aneurysms in mice. Arterioscler Thromb Vasc Biol 2014; 34:2413-20. [PMID: 25189573 DOI: 10.1161/atvbaha.114.303086] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Although nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) is reportedly essential for phagocyte host defenses, it has been found to aggravate atherosclerosis in apolipoprotein E (Apoe)-null mice through excess production of superoxide. We therefore assessed the role of NOX2 in an experimental model of abdominal aortic aneurysm (AAA) and assessed the mechanism of NOX2 action in AAA. APPROACH AND RESULTS AAA was induced in low-density lipoprotein receptor-null (Ldlr(-/-)) mice by infusing angiotensin II. Nox2 expression was elevated in the abdominal aortae of these mice during infusion of angiotensin II, with enhanced Nox2 expression mainly because of the recruitment of NOX2-enriched macrophages into AAA lesions. Unexpectedly, systemic Nox2 deficiency promoted AAA development but reduced the level of reactive oxygen species in AAA lesions. Nox2 deficiency stimulated macrophage conversion toward the M1 subset, enhancing expression of interleukin (IL)-1β and matrix metalloproteinase-9/12 mRNA. Administration of neutralizing antibody against IL-1β abolished AAA development in Nox2-deficient mice. Bone marrow transplantation experiments revealed that AAA aggravation by Nox2 deficiency is because of bone marrow-derived cells. Isolated bone marrow-derived macrophages from Nox2-null mice could not generate reactive oxygen species. In contrast, IL-1β expression in peritoneal and bone marrow-derived macrophages, but not in peritoneal neutrophils, was substantially enhanced by Nox2 deficiency. Pharmacological inhibition of Janus kinase/signal transducers and activators of transcription signaling inhibited excess IL-1β expression in Nox2-deficient macrophages, whereas matrix metalloproteinase-9 secretion was constitutively stimulated via nuclear factor-κB signals. CONCLUSIONS Nox2 deficiency enhances macrophage secretion of IL-1β and matrix metalloproteinase-9, disrupting tissue-remodeling functions in AAA lesions. These actions are unfavorable if NOX2 is to serve as a molecular target for AAA.
Collapse
Affiliation(s)
- Yasuyoshi Kigawa
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Takuro Miyazaki
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.).
| | - Xiao-Feng Lei
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Tomoya Nakamachi
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Tatsunori Oguchi
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Joo-ri Kim-Kaneyama
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Matsuo Taniyama
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Shohko Tsunawaki
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Seiji Shioda
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| | - Akira Miyazaki
- From the Department of Biochemistry (Y.K., T.M., X.-F.L., T.O., J.-r.K.-K., S.T., A.M.) and Department of Anatomy (T.N., S.S.), Showa University School of Medicine, Tokyo, Japan; Center for Biotechnology, Showa University, Tokyo, Japan (T.N.); Division of Endocrinology and Metabolism, Showa University Fujigaoka Hospital, Yokohama, Kanagawa, Japan (Y.K., M.T.); and Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan (T.N.)
| |
Collapse
|
48
|
Wilcox RA. Cutaneous T-cell lymphoma: 2014 update on diagnosis, risk-stratification, and management. Am J Hematol 2014; 89:837-51. [PMID: 25042790 DOI: 10.1002/ajh.23756] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 04/29/2014] [Indexed: 12/12/2022]
Abstract
DISEASE OVERVIEW Cutaneous T-cell lymphomas are a heterogenous group of T-cell lymphoproliferative disorders involving the skin, the majority of which may be classified as Mycosis Fungoides (MF) or Sézary Syndrome (SS). DIAGNOSIS The diagnosis of MF or SS requires the integration of clinical and histopathologic data. RISK-ADAPTED THERAPY TNMB (tumor, node, metastasis, and blood) staging remains the most important prognostic factor in MF/SS and forms the basis for a "risk-adapted," multidisciplinary approach to treatment. For patients with disease limited to the skin, expectant management or skin-directed therapies is preferred, as both disease-specific and overall survival for these patients is favorable. In contrast, patients with advanced-stage disease with significant nodal, visceral or blood involvement are generally approached with biologic-response modifiers or histone deacetylase inhibitors prior to escalating therapy to include systemic, single-agent chemotherapy. Multiagent chemotherapy (e.g., CHOP) may be employed for those patients with extensive visceral involvement requiring rapid disease control. In highly selected patients, allogeneic stem-cell transplantation may be considered.
Collapse
Affiliation(s)
- Ryan A. Wilcox
- Division of Hematology/Oncology; University of Michigan Cancer Center; Ann Arbor Michigan
| |
Collapse
|
49
|
Cross-talk between KLF4 and STAT3 regulates axon regeneration. Nat Commun 2014; 4:2633. [PMID: 24129709 PMCID: PMC3867821 DOI: 10.1038/ncomms3633] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 09/18/2013] [Indexed: 11/08/2022] Open
Abstract
Cytokine-induced activation of signal transducer and activator of transcription 3 (STAT3) promotes the regrowth of damaged axons in the adult central nervous system (CNS). Here we show that KLF4 physically interacts with STAT3 upon cytokine-induced phosphorylation of tyrosine 705 (Y705) on STAT3. This interaction suppresses STAT3-dependent gene expression by blocking its DNA-binding activity. The deletion of KLF4 in vivo induces axon regeneration of adult retinal ganglion cells (RGCs) via Janus kinase (JAK)-STAT3 signalling. This regeneration can be greatly enhanced by exogenous cytokine treatment, or removal of an endogenous JAK-STAT3 pathway inhibitor called suppressor of cytokine signalling 3 (SOCS3). These findings reveal an unexpected cross-talk between KLF4 and activated STAT3 in the regulation of axon regeneration that might have therapeutic implications in promoting repair of injured adult CNS.
Collapse
|
50
|
Xiong A, Yang Z, Shen Y, Zhou J, Shen Q. Transcription Factor STAT3 as a Novel Molecular Target for Cancer Prevention. Cancers (Basel) 2014; 6:926-57. [PMID: 24743778 PMCID: PMC4074810 DOI: 10.3390/cancers6020926] [Citation(s) in RCA: 228] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 03/11/2014] [Accepted: 03/18/2014] [Indexed: 12/11/2022] Open
Abstract
Signal Transducers and Activators of Transcription (STATs) are a family of transcription factors that regulate cell proliferation, differentiation, apoptosis, immune and inflammatory responses, and angiogenesis. Cumulative evidence has established that STAT3 has a critical role in the development of multiple cancer types. Because it is constitutively activated during disease progression and metastasis in a variety of cancers, STAT3 has promise as a drug target for cancer therapeutics. Recently, STAT3 was found to have an important role in maintaining cancer stem cells in vitro and in mouse tumor models, suggesting STAT3 is integrally involved in tumor initiation, progression and maintenance. STAT3 has been traditionally considered as nontargetable or undruggable, and the lag in developing effective STAT3 inhibitors contributes to the current lack of FDA-approved STAT3 inhibitors. Recent advances in cancer biology and drug discovery efforts have shed light on targeting STAT3 globally and/or specifically for cancer therapy. In this review, we summarize current literature and discuss the potential importance of STAT3 as a novel target for cancer prevention and of STAT3 inhibitors as effective chemopreventive agents.
Collapse
Affiliation(s)
- Ailian Xiong
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Zhengduo Yang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Yicheng Shen
- College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Qiang Shen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|