1
|
Hamade DF, Epperly MW, Fisher R, Hou W, Shields D, van Pijkeren JP, Leibowitz BJ, Coffman LG, Wang H, Huq MS, Huang Z, Rogers CJ, Vlad AM, Greenberger JS, Mukherjee A. Genetically Engineered Probiotic Limosilactobacillus reuteri Releasing IL-22 (LR-IL-22) Modifies the Tumor Microenvironment, Enabling Irradiation in Ovarian Cancer. Cancers (Basel) 2024; 16:474. [PMID: 38339228 PMCID: PMC10854600 DOI: 10.3390/cancers16030474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
Despite recent advances in cancer therapy, ovarian cancer remains the most lethal gynecological cancer worldwide, making it crucial and of the utmost importance to establish novel therapeutic strategies. Adjuvant radiotherapy has been assessed historically, but its use was limited by intestinal toxicity. We recently established the role of Limosilactobacillus reuteri in releasing IL-22 (LR-IL-22) as an effective radiation mitigator, and we have now assessed its effect in an ovarian cancer mouse model. We hypothesized that an LR-IL-22 gavage would enable intestinal radioprotection by modifying the tumor microenvironment and, subsequently, improving overall survival in female C57BL/6MUC-1 mice with widespread abdominal syngeneic 2F8cis ovarian cancer. Herein, we report that the LR-IL-22 gavage not only improved overall survival in mice when combined with a PD-L1 inhibitor by inducing differential gene expression in irradiated stem cells but also induced PD-L1 protein expression in ovarian cancer cells and mobilized CD8+ T cells in whole abdomen irradiated mice. The addition of LR-IL-22 to a combined treatment modality with fractionated whole abdomen radiation (WAI) and systemic chemotherapy and immunotherapy regimens can facilitate a safe and effective protocol to reduce tumor burden, increase survival, and improve the quality of life of a locally advanced ovarian cancer patient.
Collapse
Affiliation(s)
- Diala F. Hamade
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Michael W. Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Wen Hou
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Donna Shields
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | | | - Brian J. Leibowitz
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Lan G. Coffman
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15260, USA; (H.W.); (Z.H.)
| | - M. Saiful Huq
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Ziyu Huang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15260, USA; (H.W.); (Z.H.)
| | | | - Anda M. Vlad
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD 20850, USA;
| | - Joel S. Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| | - Amitava Mukherjee
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (D.F.H.); (M.W.E.); (R.F.); (W.H.); (D.S.); (B.J.L.); (M.S.H.); (J.S.G.)
| |
Collapse
|
2
|
Xu X, Sun R, Li Y, Wang J, Zhang M, Xiong X, Xie D, Jin X, Zhao M. Comprehensive bioinformatic analysis of the expression and prognostic significance of TSC22D domain family genes in adult acute myeloid leukemia. BMC Med Genomics 2023; 16:117. [PMID: 37237254 DOI: 10.1186/s12920-023-01550-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND TSC22D domain family genes, including TSC22D1-4, play a principal role in cancer progression. However, their expression profiles and prognostic significance in adult acute myeloid leukemia (AML) remain unknown. METHODS The online databases, including HPA, CCLE, EMBL-EBI, GEPIA2, BloodSpot, GENT2, UCSCXenaShiny, GSCALite, cBioportal, and GenomicScape, utilized the data of TCGA and GEO to investigate gene expression, mutation, copy number variation (CNV), and prognostic significance of the TSC22D domain family in adult AML. Computational analysis of resistance (CARE) was used to explore the effect of TSC22D3 expression on drug response. Functional enrichment analysis of TSC22D3 was performed in the TRRUST Version 2 database. The STRING, Pathway Commons, and AnimalTFDB3.0 databases were used to investigate the protein-protein interaction (PPI) network of TSC22D3. Harmonizome was used to predict target genes and kinases regulated by TSC22D3. The StarBase v2.0 and CancermiRNome databases were used to predict miRNAs regulated by TSC22D3. UCSCXenaShiny was used to investigate the correlation between TSC22D3 expression and immune infiltration. RESULTS Compared with normal adult hematopoietic stem cells (HSCs), the expression of TSC22D3 and TSC22D4 in adult AML tissues was markedly up-regulated, whereas TSC22D1 expression was markedly down-regulated. The expression of TSC22D1 and TSC22D3 was significantly increased in adult AML tissues compared to normal adult tissues. High TSC22D3 expression was significantly associated with poor overall survival (OS) and event-free survival (EFS) in adult AML patients. Univariate and multivariate Cox analysis showed that overexpression of TSC22D3 was independently associated with adverse OS of adult AML patients. High TSC22D3 expression had a adverse impact on OS and EFS of adult AML patients in the chemotherapy group. TSC22D3 expression correlated with drug resistance to BCL2 inhibitors. Functional enrichment analysis indicated that TSC22D3 might promote AML progression. MIR143-3p sponging TSC22D3 might have anti-leukemia effect in adult AML. CONCLUSIONS A significant increase in TSC22D3 expression was observed in adult AML tissues compared to normal adult HSCs and tissues. The prognosis of adult AML patients with high TSC22D3 expression was unfavorable, which could severe as a new prognostic biomarker and potential target for adult AML.
Collapse
Affiliation(s)
- XiaoQiang Xu
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300192, China
- Department of Hematology, Shanxi Fenyang Hospital, Fenyang, 032200, China
| | - Rui Sun
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - YuanZhang Li
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300192, China
| | - JiaXi Wang
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300192, China
| | - Meng Zhang
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300192, China
| | - Xia Xiong
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300192, China
| | - DanNi Xie
- The First Central Clinical School, Tianjin Medical University, Tianjin, 300192, China
| | - Xin Jin
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300192, China
| | - MingFeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300192, China.
| |
Collapse
|
3
|
Lee SH, Choi D. Transforming Stimulated Clone 22 (TSC-22) Interacts Directly with Bromodomain-Containing Protein 7 (BRD7) to Enhance the Inhibition of Extracellular Signal-Regulate Kinase (ERK) Pathway in Ovarian Cancer. Dev Reprod 2022; 26:117-126. [PMID: 36285148 PMCID: PMC9578317 DOI: 10.12717/dr.2022.26.3.117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/08/2022] [Accepted: 09/02/2022] [Indexed: 11/06/2022]
Abstract
Bromodomain-containing protein 7 (BRD7) participates in many cellular processes
and embryo development. BRD7 is down-regulated in various cancers and evidence
of its tumor suppressor function has been accumulating. Here, we identified
transforming stimulated clone 22 (TSC-22) as a novel BRD7 interacting protein
and show its novel function as a positive regulator of BRD7. We found that
TSC-22 expression potentiated the inactivation of the extracellular
signal-regulate kinase (ERK) pathway by BRD7. Our data establishes TSC-22 as a
modulator of BRD7 and unravels the molecular mechanisms that drive the
synergistic tumor-suppressing effects of TSC-22 and BRD7. Our findings may open
new avenues for developing novel molecular therapies for tumors exhibiting
down-regulated BRD7 and/or TSC-22.
Collapse
Affiliation(s)
- Seung-Hoon Lee
- Department of Life Science, YongIn
University, Yongin 17092, Korea,Corresponding author Seung-Hoon
Lee, Department of Life Science, YongIn University, Yongin 17092, Korea. Tel:
+82-31-8020-2780, E-mail:
| | - Donchan Choi
- Department of Life Science, YongIn
University, Yongin 17092, Korea
| |
Collapse
|
4
|
Gibriel AA, Ismail MF, Sleem H, Zayed N, Yosry A, El-Nahaas SM, Shehata NI. Diagnosis and staging of HCV associated fibrosis, cirrhosis and hepatocellular carcinoma with target identification for miR-650, 552-3p, 676-3p, 512-5p and 147b. Cancer Biomark 2022; 34:413-430. [DOI: 10.3233/cbm-210456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND: Chronic HCV infection progresses to fibrosis, cirrhosis and hepatocellular carcinoma (HCC). The latter represents the third most common cause for cancer mortality. Currently, there is no reliable non-invasive biomarker for diagnosis of HCV mediated disorders. OBJECTIVE: Profiling expression signature for circulatory miRNAs in the plasma of 167 Egyptian patients (40 healthy, 48 HCV fibrotic, 39 HCV cirrhotic and 40 HCV-HCC cases). METHODS: QRTPCR was used to quantify expression signature for circulatory miRNAs. RESULTS: MiR-676 and miR-650 were powerful in discriminating cirrhotic and late fibrosis from HCC. MiR-650 could distinguish mild (f0-f1) and advanced (f2-f3) fibrosis from HCC cases. MiR-650 and miR-147b could distinguish early fibrosis from healthy controls meanwhile miR-676 and miR-147b could effectively distinguish between mild chronic and (f1-f3) cases from healthy individuals. All studied miRNAs, except miR-512, can differentiate between (f0-f3) cases and healthy controls. Multivariate logistic regression revealed three potential miRNA panels for effective differentiation of HCC, cirrhotic and chronic liver cases. MiR-676-3p and miR-512-5p were significantly correlated in (f1-f3) fibrosis meanwhile miR-676 and miR-512 could differentiate between cirrhosis and (f0-f3) cases. Both miR-650 and miR-512-5p were positively correlated in the cirrhotic group and in (f0-f4) group. Putative targets for investigated miRNAs were also determined. CONCLUSIONS: Investigated miRNAs could assist in staging and diagnosis of HCV associated disorders.
Collapse
Affiliation(s)
- Abdullah Ahmed Gibriel
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, Egypt
| | - Manal Fouad Ismail
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hameis Sleem
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, Egypt
| | - Naglaa Zayed
- Endemic Medicine Department and Hepatology Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ayman Yosry
- Endemic Medicine Department and Hepatology Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Saeed M. El-Nahaas
- Endemic Medicine Department and Hepatology Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
5
|
Kamimura R, Uchida D, Kanno SI, Shiraishi R, Hyodo T, Sawatani Y, Shimura M, Hasegawa T, Tsubura-Okubo M, Yaguchi E, Komiyama Y, Fukumoto C, Izumi S, Fujita A, Wakui T, Kawamata H. Identification of Binding Proteins for TSC22D1 Family Proteins Using Mass Spectrometry. Int J Mol Sci 2021; 22:ijms222010913. [PMID: 34681573 PMCID: PMC8536140 DOI: 10.3390/ijms222010913] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 11/29/2022] Open
Abstract
TSC-22 (TGF-β stimulated clone-22) has been reported to induce differentiation, growth inhibition, and apoptosis in various cells. TSC-22 is a member of a family in which many proteins are produced from four different family genes. TSC-22 (corresponding to TSC22D1-2) is composed of 144 amino acids translated from a short variant mRNA of the TSC22D1 gene. In this study, we attempted to determine the intracellular localizations of the TSC22D1 family proteins (TSC22D1-1, TSC-22 (TSC22D1-2), and TSC22(86) (TSC22D1-3)) and identify the binding proteins for TSC22D1 family proteins by mass spectrometry. We determined that TSC22D1-1 was mostly localized in the nucleus, TSC-22 (TSC22D1-2) was localized in the cytoplasm, mainly in the mitochondria and translocated from the cytoplasm to the nucleus after DNA damage, and TSC22(86) (TSC22D1-3) was localized in both the cytoplasm and nucleus. We identified multiple candidates of binding proteins for TSC22D1 family proteins in in vitro pull-down assays and in vivo binding assays. Histone H1 bound to TSC-22 (TSC22D1-2) or TSC22(86) (TSC22D1-3) in the nucleus. Guanine nucleotide-binding protein-like 3 (GNL3), which is also known as nucleostemin, bound to TSC-22 (TSC22D1-2) in the nucleus. Further investigation of the interaction of the candidate binding proteins with TSC22D1 family proteins would clarify the biological roles of TSC22D1 family proteins in several cell systems.
Collapse
Affiliation(s)
- Ryouta Kamimura
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Daisuke Uchida
- Department of Oral and Maxillofacial Surgery, Ehime University Graduate School of Medicine, Shitsukawa, Toon 791-0295, Ehime, Japan;
| | - Shin-ichiro Kanno
- Division of Dynamic Proteome, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-machi, Sendai 980-8575, Aobaku, Japan;
| | - Ryo Shiraishi
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Toshiki Hyodo
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Yuta Sawatani
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
- Section of Dentistry, Oral and Maxillofacial Surgery, Kamitsuga General Hospital, 1-1033 Shimoda-machi, Kanuma 322-8550, Tochigi, Japan
| | - Michiko Shimura
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
- Section of Dentistry and Oral and Maxillofacial Surgery, Sano Kosei General Hospital, 1728 Horigomecho, Sano 327-8511, Tochigi, Japan
| | - Tomonori Hasegawa
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Maki Tsubura-Okubo
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
- Section of Dentistry and Oral and Maxillofacial Surgery, Sano Kosei General Hospital, 1728 Horigomecho, Sano 327-8511, Tochigi, Japan
| | - Erika Yaguchi
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Yuske Komiyama
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Chonji Fukumoto
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Sayaka Izumi
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Atsushi Fujita
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Takahiro Wakui
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
| | - Hitoshi Kawamata
- Department of Oral and Maxillofacial Surgery, Dokkyo Medical University School of Medicine, 880 Kita-kobayashi, Shimotsuga, Mibu 321-0293, Tochigi, Japan; (R.K.); (R.S.); (T.H.); (Y.S.); (M.S.); (T.H.); (M.T.-O.); (E.Y.); (Y.K.); (C.F.); (S.I.); (A.F.); (T.W.)
- Correspondence: ; Tel.: +81-282-87-2130; Fax: +81-282-86-1681
| |
Collapse
|
6
|
Qin S, Li B, Li R, Cai Y, Zheng K, Huang H, Xiao F, Zeng M, Xu X. Proteomic characteristics and identification of PM 2.5-induced differentially expressed proteins in hepatocytes and c-Myc silenced hepatocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 209:111838. [PMID: 33387776 DOI: 10.1016/j.ecoenv.2020.111838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/16/2020] [Accepted: 12/19/2020] [Indexed: 06/12/2023]
Abstract
Proteomics and bioinformatics were applied to explore PM2.5-induced differentially expressed proteins (DEPs) in hepatocytes (L02 cells) and c-Myc-silenced hepatocytes. L02 cells and c-Myc-silenced hepatocytes were treated with PM2.5 for 24 h. Fifty-two DEPs were screened in L02 hepatocytes, of which 28 were upregulated and 24 were downregulated. Forty-one DEPs were screened in the c-Myc-silenced hepatocytes, of which 31 were upregulated and 10 were downregulated. GO analysis showed that DEPs in L02 cells were mainly concentrated in the cytosol and were involved in biological processes such as the response to metal ions. DEPs in c-Myc-silenced cells were mainly enriched in the extracellular space and were involved in lipoprotein metabolism. KEGG analysis showed that DEPs in L02 cells were mainly involved in arachidonic acid metabolism and mineral absorption. DEPs in c-Myc-silenced cells were mainly enriched in pathways involving nerve absorption, complement and coagulation cascades, and other pathways. Twenty key proteins, including Metallothionein-2A (MT2A), Metallothionein-1X (MT1X), zinc transporter ZIP10 (SLC39A10) and Serine protease 23 (PRSS23) were screened in two groups through analysis of protein-protein interactions. Based on the identification of the selected DEPs, PRSS23 and SLC39A10 might be the potential biomarker of PM2.5-induced carcinogenesis, which provide the scientific basis for further research into the carcinogenic mechanisms of PM2.5.
Collapse
Affiliation(s)
- Shuangjian Qin
- Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China; Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China
| | - Boru Li
- Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China; Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China
| | - Runbing Li
- Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China; School of public health, University of South China, Hengyang, Hunan 421001, China
| | - Ying Cai
- Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China; School of public health, University of South China, Hengyang, Hunan 421001, China
| | - Kai Zheng
- Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China; School of public health, University of South China, Hengyang, Hunan 421001, China
| | - Haiyan Huang
- Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China
| | - Fang Xiao
- Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China.
| | - Ming Zeng
- Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China.
| | - Xinyun Xu
- Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
7
|
Boeckx B, Shahi RB, Smeets D, De Brakeleer S, Decoster L, Van Brussel T, Galdermans D, Vercauter P, Decoster L, Alexander P, Berchem G, Ocak S, Vuylsteke P, Deschepper K, Lambrechts M, Cappoen N, Teugels E, Lambrechts D, De Greve J. The genomic landscape of nonsmall cell lung carcinoma in never smokers. Int J Cancer 2020; 146:3207-3218. [DOI: 10.1002/ijc.32797] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 10/11/2019] [Accepted: 10/15/2019] [Indexed: 01/30/2023]
Affiliation(s)
- Bram Boeckx
- Laboratory of Translational GeneticsVIB Center for Cancer Biology, VIB Leuven Belgium
- Laboratory of Translational Genetics, Department of Human GeneticsUniversity of Leuven (KULeuven) Leuven Belgium
| | - Rajendra B. Shahi
- Laboratory of Medical and Molecular Oncology (LMMO), Department of Medical Oncology, Oncologisch Centrum, UZ BrusselVrije Universiteit Brussel (VUB) Brussels Belgium
| | - Dominiek Smeets
- Laboratory of Translational GeneticsVIB Center for Cancer Biology, VIB Leuven Belgium
- Laboratory of Translational Genetics, Department of Human GeneticsUniversity of Leuven (KULeuven) Leuven Belgium
| | - Sylvia De Brakeleer
- Laboratory of Medical and Molecular Oncology (LMMO), Department of Medical Oncology, Oncologisch Centrum, UZ BrusselVrije Universiteit Brussel (VUB) Brussels Belgium
| | - Lore Decoster
- Laboratory of Medical and Molecular Oncology (LMMO), Department of Medical Oncology, Oncologisch Centrum, UZ BrusselVrije Universiteit Brussel (VUB) Brussels Belgium
| | - Thomas Van Brussel
- Laboratory of Translational GeneticsVIB Center for Cancer Biology, VIB Leuven Belgium
- Laboratory of Translational Genetics, Department of Human GeneticsUniversity of Leuven (KULeuven) Leuven Belgium
| | | | | | - Lynn Decoster
- Department of PulmonologyAZ Turnhout Turnhout Belgium
| | | | - Guy Berchem
- Centre Hospitalier de Luxembourg Luxembourg City Luxemburg
| | - Sebahat Ocak
- CHU UCL Namur (Godinne Site) Yvoir Belgium
- Institut de Recherche Expérimentale et Clinique (IREC), Pneumology Pole, Université Catholique de Louvain (UCL) Ottignies‐Louvain‐la‐Neuve Belgium
| | - Peter Vuylsteke
- Université Catholique de Louvain, CHU UCL Namur, Site Sainte Elisabeth Namur Belgium
| | | | | | - Nadia Cappoen
- Laboratory of Medical and Molecular Oncology (LMMO), Department of Medical Oncology, Oncologisch Centrum, UZ BrusselVrije Universiteit Brussel (VUB) Brussels Belgium
| | - Erik Teugels
- Laboratory of Medical and Molecular Oncology (LMMO), Department of Medical Oncology, Oncologisch Centrum, UZ BrusselVrije Universiteit Brussel (VUB) Brussels Belgium
| | - Diether Lambrechts
- Laboratory of Translational GeneticsVIB Center for Cancer Biology, VIB Leuven Belgium
- Laboratory of Translational Genetics, Department of Human GeneticsUniversity of Leuven (KULeuven) Leuven Belgium
| | - Jacques De Greve
- Laboratory of Medical and Molecular Oncology (LMMO), Department of Medical Oncology, Oncologisch Centrum, UZ BrusselVrije Universiteit Brussel (VUB) Brussels Belgium
| |
Collapse
|
8
|
Talreja J, Bauerfeld C, Sendler E, Pique-Regi R, Luca F, Samavati L. Derangement of Metabolic and Lysosomal Gene Profiles in Response to Dexamethasone Treatment in Sarcoidosis. Front Immunol 2020; 11:779. [PMID: 32477331 PMCID: PMC7235403 DOI: 10.3389/fimmu.2020.00779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/06/2020] [Indexed: 12/14/2022] Open
Abstract
Glucocorticoids (GCs) play a central role in modulation of inflammation in various diseases, including respiratory diseases such as sarcoidosis. Surprisingly, the specific anti-inflammatory effects of GCs on different myeloid cells especially in macrophages remain poorly understood. Sarcoidosis is a systemic granulomatous disease of unknown etiology that occurs worldwide and is characterized by granuloma formation in different organs. Alveolar macrophages play a role in sarcoidosis granuloma formation and progressive lung disease. The goal of the present study is to identify the effect of GCs on transcriptomic profiles and the cellular pathways in sarcoidosis alveolar macrophages and their corresponding blood myeloid cells. We determined and compared the whole transcriptional signatures of alveolar macrophages from sarcoidosis patients and blood CD14+ monocytes of the same subjects in response to in vitro treatment with dexamethasone (DEX) via RNA-sequencing. In response to DEX, we identified 2,834 genes that were differentially expressed in AM. Predominant pathways affected were as following: metabolic pathway (FDR = 4.1 × 10−10), lysosome (FDR = 6.3 × 10−9), phagosome (FDR = 3.9 × 10−5). The DEX effect on AMs is associated with metabolic derangements involving glycolysis, oxidative phosphorylation and lipid metabolisms. In contrast, the top impacted pathways in response to DEX treatment in blood CD14+ monocytes were as following; cytokine-cytokine receptor interaction (FDR = 6 × 10−6) and transcriptional misregulation in cancer (FDR = 1 × 10−4). Pathways similarly affected in both cell types were genes involved in lysosomes, cytoskeleton and transcriptional misregulation in cancer. These data suggest that the different effects of DEX on AMs and peripheral blood monocytes are partly dictated by lineage specific transcriptional programs and their physiological functions.
Collapse
Affiliation(s)
- Jaya Talreja
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine and Detroit Medical Center, Wayne State University, Detroit, MI, United States
| | - Christian Bauerfeld
- Division of Critical Care, Department of Pediatrics, School of Medicine and Detroit Medical Center, Wayne State University, Detroit, MI, United States
| | - Edward Sendler
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, United States
| | - Roger Pique-Regi
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, United States.,Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, United States
| | - Francesca Luca
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, United States.,Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, United States
| | - Lobelia Samavati
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine and Detroit Medical Center, Wayne State University, Detroit, MI, United States.,Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, United States
| |
Collapse
|
9
|
La HM, Chan AL, Legrand JMD, Rossello FJ, Gangemi CG, Papa A, Cheng Q, Morand EF, Hobbs RM. GILZ-dependent modulation of mTORC1 regulates spermatogonial maintenance. Development 2018; 145:dev.165324. [PMID: 30126904 DOI: 10.1242/dev.165324] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 08/06/2018] [Indexed: 12/15/2022]
Abstract
Male fertility is dependent on spermatogonial stem cells (SSCs) that self-renew and produce differentiating germ cells. Growth factors produced within the testis are essential for SSC maintenance but intrinsic factors that dictate the SSC response to these stimuli are poorly characterised. Here, we have studied the role of GILZ, a TSC22D family protein and spermatogenesis regulator, in spermatogonial function and signalling. Although broadly expressed in the germline, GILZ was prominent in undifferentiated spermatogonia and Gilz deletion in adults resulted in exhaustion of the GFRα1+ SSC-containing population and germline degeneration. GILZ loss was associated with mTORC1 activation, suggesting enhanced growth factor signalling. Expression of deubiquitylase USP9X, an mTORC1 modulator required for spermatogenesis, was disrupted in Gilz mutants. Treatment with an mTOR inhibitor rescued GFRα1+ spermatogonial failure, indicating that GILZ-dependent mTORC1 inhibition is crucial for SSC maintenance. Analysis of cultured undifferentiated spermatogonia lacking GILZ confirmed aberrant activation of ERK MAPK upstream mTORC1 plus USP9X downregulation and interaction of GILZ with TSC22D proteins. Our data indicate an essential role for GILZ-TSC22D complexes in ensuring the appropriate response of undifferentiated spermatogonia to growth factors via distinct inputs to mTORC1.
Collapse
Affiliation(s)
- Hue M La
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria 3800, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Ai-Leen Chan
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria 3800, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Julien M D Legrand
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria 3800, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Fernando J Rossello
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria 3800, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Christina G Gangemi
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria 3800, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Antonella Papa
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Qiang Cheng
- Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria 3800, Australia
| | - Eric F Morand
- Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria 3800, Australia
| | - Robin M Hobbs
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria 3800, Australia .,Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| |
Collapse
|
10
|
Low levels of TSC22 enhance tumorigenesis by inducing cell proliferation in colorectal cancer. Biochem Biophys Res Commun 2018; 497:1062-1067. [PMID: 29481799 DOI: 10.1016/j.bbrc.2018.02.177] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 02/23/2018] [Indexed: 11/21/2022]
Abstract
Transforming growth factor β-stimulated clone 22 domain 1 (TSC22) has been identified as a cancer suppressor gene in various kinds of cancers. The purpose of this study was to explore the expression of TSC22 in colorectal cancer (CRC) tissues and cell lines. 24 matched CRC and normal tissue samples by qPCR along with 18 pairs of them by Western blot demonstrated TSC22 level was decreased in CRC compared with normal tissue. The protein expression of TSC22 was examined in 310 CRC specimens. Results showed low expression of TSC22 was significantly correlated with tumor size (P = 0.048) and tumor infiltration (P = 0.016). Kaplan-Meier method suggested low expression of TSC22 was inversely associated with OS for 276 samples (P < 0.01). Multivariate Cox regression analysis confirmed TSC22 expression as independent predictors of the OS in CRC patients. Furthermore, we found TSC22 could suppress tumor by inhibiting cell proliferation in CRC cell lines.
Collapse
|
11
|
Zheng L, Suzuki H, Nakajo Y, Nakano A, Kato M. Regulation of c-MYC transcriptional activity by transforming growth factor-beta 1-stimulated clone 22. Cancer Sci 2018; 109:395-402. [PMID: 29224245 PMCID: PMC5797808 DOI: 10.1111/cas.13466] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/27/2017] [Accepted: 11/30/2017] [Indexed: 12/20/2022] Open
Abstract
c‐MYC stimulates cell proliferation through the suppression of cyclin‐dependent kinase (CDK) inhibitors including P15 (CDKN2B) and P21 (CDKN1A). It also activates E‐box‐mediated transcription of various target genes including telomerase reverse transcriptase (TERT) that is involved in cellular immortality and tumorigenesis. Transforming growth factor‐beta 1 (TGF‐β1)‐stimulated clone 22 (TSC‐22/TSC22D1) encodes a highly conserved leucine zipper protein that is induced by various stimuli, including TGF‐β. TSC‐22 inhibits cell growth in mammalian cells and in Xenopus embryos. However, underlying mechanisms of growth inhibition by TSC‐22 remain unclear. Here, we show that TSC‐22 physically interacts with c‐MYC to inhibit the recruitment of c‐MYC on the P15 (CDKN2B) and P21 (CDKN1A) promoters, effectively inhibiting c‐MYC‐mediated suppression of P15 (CDKN2B) and also P21 (CDKN1A) promoter activities. In contrast, TSC‐22 enhances c‐MYC‐mediated activation of the TERT promoter. Additionally, the expression of TSC‐22 in embryonic stem cells inhibits cell growth without affecting its pluripotency‐related gene expression. These results indicate that TSC‐22 differentially regulates c‐MYC‐mediated transcriptional activity to regulate cell proliferation.
Collapse
Affiliation(s)
- Ling Zheng
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroyuki Suzuki
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yuka Nakajo
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Akinobu Nakano
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Mitsuyasu Kato
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
12
|
Jostes S, Nettersheim D, Fellermeyer M, Schneider S, Hafezi F, Honecker F, Schumacher V, Geyer M, Kristiansen G, Schorle H. The bromodomain inhibitor JQ1 triggers growth arrest and apoptosis in testicular germ cell tumours in vitro and in vivo. J Cell Mol Med 2016; 21:1300-1314. [PMID: 28026145 PMCID: PMC5487916 DOI: 10.1111/jcmm.13059] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/15/2016] [Indexed: 12/21/2022] Open
Abstract
Type II testicular germ cell cancers (TGCT) are the most frequently diagnosed tumours in young men (20–40 years) and are classified as seminoma or non‐seminoma. TGCTs are commonly treated by orchiectomy and chemo‐ or radiotherapy. However, a subset of metastatic non‐seminomas (embryonal carcinomas) displays only incomplete remission or relapse and requires novel treatment options. Recent studies have shown effective application of the small‐molecule inhibitor JQ1 in tumour therapy, which interferes with the function of ‘bromodomain and extraterminal (BET)’ proteins. JQ1‐treated TGCT cell lines display up‐regulation of genes indicative for DNA damage and cellular stress response and induce cell cycle arrest. Embryonal carcinoma (EC) cell lines, which presented as JQ1 sensitive, display down‐regulation of pluripotency factors and induction of mesodermal differentiation. In contrast, seminoma‐like TCam‐2 cells tolerated higher JQ1 concentrations and were resistant to differentiation. ECs xenografted in vivo showed a reduction in tumour size, proliferation rate and angiogenesis in response to JQ1. Finally, the combination of JQ1 and the histone deacetylase inhibitor romidepsin allowed for lower doses and less frequent application, compared with monotherapy. Thus, we propose that JQ1 in combination with romidepsin may serve as a novel therapeutic option for (mixed) TGCTs.
Collapse
Affiliation(s)
- Sina Jostes
- Institute of Pathology, Department of Developmental Pathology, University Medical School, Bonn, Germany
| | - Daniel Nettersheim
- Institute of Pathology, Department of Developmental Pathology, University Medical School, Bonn, Germany
| | - Martin Fellermeyer
- Institute of Pathology, Department of Developmental Pathology, University Medical School, Bonn, Germany
| | - Simon Schneider
- Institute of Pathology, Department of Developmental Pathology, University Medical School, Bonn, Germany
| | - François Hafezi
- Institute of Pathology, Department of Developmental Pathology, University Medical School, Bonn, Germany
| | | | - Valerie Schumacher
- Department of Urology, Boston Children's Hospital, Boston, MA, USA.,Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Matthias Geyer
- Institute of Innate Immunity, Department of Structural Immunology, University Medical School, Bonn, Germany
| | - Glen Kristiansen
- Institute of Pathology, University Medical School, Bonn, Germany
| | - Hubert Schorle
- Institute of Pathology, Department of Developmental Pathology, University Medical School, Bonn, Germany
| |
Collapse
|
13
|
Acute transcriptional up-regulation specific to osteoblasts/osteoclasts in medaka fish immediately after exposure to microgravity. Sci Rep 2016; 6:39545. [PMID: 28004797 PMCID: PMC5177882 DOI: 10.1038/srep39545] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/24/2016] [Indexed: 12/22/2022] Open
Abstract
Bone loss is a serious problem in spaceflight; however, the initial action of microgravity has not been identified. To examine this action, we performed live-imaging of animals during a space mission followed by transcriptome analysis using medaka transgenic lines expressing osteoblast and osteoclast-specific promoter-driven GFP and DsRed. In live-imaging for osteoblasts, the intensity of osterix- or osteocalcin-DsRed fluorescence in pharyngeal bones was significantly enhanced 1 day after launch; and this enhancement continued for 8 or 5 days. In osteoclasts, the signals of TRAP-GFP and MMP9-DsRed were highly increased at days 4 and 6 after launch in flight. HiSeq from pharyngeal bones of juvenile fish at day 2 after launch showed up-regulation of 2 osteoblast- and 3 osteoclast- related genes. Gene ontology analysis for the whole-body showed that transcription of genes in the category “nucleus” was significantly enhanced; particularly, transcription-regulators were more up-regulated at day 2 than at day 6. Lastly, we identified 5 genes, c-fos, jun-B-like, pai-1, ddit4 and tsc22d3, which were up-regulated commonly in the whole-body at days 2 and 6, and in the pharyngeal bone at day 2. Our results suggested that exposure to microgravity immediately induced dynamic alteration of gene expression levels in osteoblasts and osteoclasts.
Collapse
|
14
|
Ekim Üstünel B, Friedrich K, Maida A, Wang X, Krones-Herzig A, Seibert O, Sommerfeld A, Jones A, Sijmonsma TP, Sticht C, Gretz N, Fleming T, Nawroth PP, Stremmel W, Rose AJ, Berriel-Diaz M, Blüher M, Herzig S. Control of diabetic hyperglycaemia and insulin resistance through TSC22D4. Nat Commun 2016; 7:13267. [PMID: 27827363 PMCID: PMC5105165 DOI: 10.1038/ncomms13267] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/15/2016] [Indexed: 12/29/2022] Open
Abstract
Obesity-related insulin resistance represents the core component of the metabolic syndrome, promoting glucose intolerance, pancreatic beta cell failure and type 2 diabetes. Efficient and safe insulin sensitization and glucose control remain critical therapeutic aims to prevent diabetic late complications Here, we identify transforming growth factor beta-like stimulated clone (TSC) 22 D4 as a molecular determinant of insulin signalling and glucose handling. Hepatic TSC22D4 inhibition both prevents and reverses hyperglycaemia, glucose intolerance and insulin resistance in diabetes mouse models. TSC22D4 exerts its effects on systemic glucose homeostasis—at least in part—through the direct transcriptional regulation of the small secretory protein lipocalin 13 (LCN13). Human diabetic patients display elevated hepatic TSC22D4 expression, which correlates with decreased insulin sensitivity, hyperglycaemia and LCN13 serum levels. Our results establish TSC22D4 as a checkpoint in systemic glucose metabolism in both mice and humans, and propose TSC22D4 inhibition as an insulin sensitizing option in diabetes therapy. TSC22D4 regulates hepatic lipoprotein production, but has so far mainly been studied in the context of cancer cachexia. Here, the authors show TSC22D4 inhibition improves insulin sensitivity in several mouse models of diabetes, which they attribute at least in part to the induction of secreted LCN13.
Collapse
Affiliation(s)
- Bilgen Ekim Üstünel
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Kilian Friedrich
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany.,Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Adriano Maida
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Xiaoyue Wang
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Anja Krones-Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Oksana Seibert
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Anke Sommerfeld
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Allan Jones
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Tjeerd P Sijmonsma
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Carsten Sticht
- Center for Clinical Research, Medical Faculty Mannheim, 68167 Mannheim, Germany
| | - Norbert Gretz
- Center for Clinical Research, Medical Faculty Mannheim, 68167 Mannheim, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, Heidelberg University, 69120 Heidelberg, Germany
| | - Peter P Nawroth
- Department of Medicine I and Clinical Chemistry, Heidelberg University, 69120 Heidelberg, Germany
| | - Wolfgang Stremmel
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Adam J Rose
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Mauricio Berriel-Diaz
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, 85764 Neuherberg, Germany
| |
Collapse
|
15
|
Liang F, Li Q, Li X, Li Z, Gong Z, Deng H, Xiang B, Zhou M, Li X, Li G, Zeng Z, Xiong W. TSC22D2 interacts with PKM2 and inhibits cell growth in colorectal cancer. Int J Oncol 2016; 49:1046-56. [PMID: 27573352 DOI: 10.3892/ijo.2016.3599] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/16/2016] [Indexed: 11/06/2022] Open
Abstract
We previously identified TSC22D2 (transforming growth factor β-stimulated clone 22 domain family, member 2) as a novel cancer-associated gene in a rare multi-cancer family. However, its role in tumor development remains completely unknown. In this study, we found that TSC22D2 was significantly downregulated in colorectal cancer (CRC) and that TSC22D2 overexpression inhibited cell growth. Using a co-immunoprecipitation (co-IP) assay combined with mass spectrometry analysis to identify TSC22D2-interacting proteins, we demonstrated that TSC22D2 interacts with pyruvate kinase isoform M2 (PKM2). These findings were confirmed by the results of immunoprecipitation and immunofluorescence assays. Moreover, overexpression of TSC22D2 reduced the level of nuclear PKM2 and suppressed cyclin D1 expression. Collectively, our study reveals a growth suppressor function of TSC22D2 that is at least partially dependent on the TSC22D2-PKM2-cyclinD1 regulatory axis. In addition, our data provide important clues that might contribute to future studies evaluating the role of TSC22D2.
Collapse
Affiliation(s)
- Fang Liang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, Hunan, P.R. China
| | - Qiao Li
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, Hunan, P.R. China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Zheng Li
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, Hunan, P.R. China
| | - Zhaojian Gong
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health and The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, P.R. China
| | - Hao Deng
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Bo Xiang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, Hunan, P.R. China
| | - Ming Zhou
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, Hunan, P.R. China
| | - Xiaoling Li
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, Hunan, P.R. China
| | - Guiyuan Li
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, Hunan, P.R. China
| | - Zhaoyang Zeng
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, Hunan, P.R. China
| | - Wei Xiong
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Cancer Research Institute, Central South University, Changsha, Hunan, P.R. China
| |
Collapse
|
16
|
Li Q, Chen P, Zeng Z, Liang F, Song Y, Xiong F, Li X, Gong Z, Zhou M, Xiang B, Peng C, Li X, Chen X, Li G, Xiong W. Yeast two-hybrid screening identified WDR77 as a novel interacting partner of TSC22D2. Tumour Biol 2016; 37:12503-12512. [PMID: 27337956 DOI: 10.1007/s13277-016-5113-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/09/2016] [Indexed: 12/14/2022] Open
Abstract
Transforming growth factor β-stimulated clone 22 domain family, member 2 (TSC22D2), a member of the TSC22D family, has been implicated as a tumor-associated gene, but its function remains unknown. To further explore its biological role, yeast two-hybrid screening combined with multiple bioinformatics tools was used to identify 44 potential interacting partners of the TSC22D2 protein that were mainly involved in gene transcription, cellular metabolism, and cell cycle regulation. The protein WD repeat domain 77 (WDR77) was selected for further validation due to its function in the cell cycle and tumor development, as well as its high detection frequency in the yeast two-hybrid assay. Immunoprecipitation and immunofluorescence experiments confirmed an interaction between the TSC22D2 and WDR77 proteins. Our work greatly expands the putative protein interaction network of TSC22D2 and provides deeper insight into the biological functions of the TSC22D2 and WDR77 proteins.
Collapse
Affiliation(s)
- Qiao Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Pan Chen
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| | - Fang Liang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yali Song
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Fang Xiong
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaojian Gong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Ming Zhou
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bo Xiang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Cong Peng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiang Chen
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
17
|
Overexpression of TSC-22 (transforming growth factor- β-stimulated clone-22) causes marked obesity, splenic abnormality and B cell lymphoma in transgenic mice. Oncotarget 2016; 7:14310-23. [PMID: 26872059 PMCID: PMC4924717 DOI: 10.18632/oncotarget.7308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/29/2016] [Indexed: 11/30/2022] Open
Abstract
In this study, we generated transgenic (Tg) mice, which overexpressed transforming growth factor (TGF)-β stimulated clone-22 (TSC-22), and investigate the functional role of TSC-22 on their development and pathogenesis. We obtained 13 Tg-founders (two mice from C57BL6/J and 11 mice from BDF1). Three of 13 Tg-founders were sterile, and the remaining Tg-founders also could generate only a limited number of the F1 generation. We obtained 32 Tg-F1 mice. Most of the Tg-mice showed marked obesity. Histopathological examination could be performed on 31 Tg-mice; seventeen mice died by some disease in their entire life and 14 mice were killed for examination. Most of the Tg-mice examined showed splenic abnormality, in which marked increase of the megakaryocytes, unclearness of the margin of the red pulp and the white pulp, and the enlargement of the white pulp was observed. B cell lymphoma was developed in 10 (71%) of 14 disease-died F1 mice. These results indicate that constitutive over-expression of TSC-22 might disturb the normal embryogenesis and the normal lipid metabolism, and induce the oncogenic differentiation of hematopoietic cells.
Collapse
|
18
|
Pépin A, Biola-Vidamment A, Latré de Laté P, Espinasse MA, Godot V, Pallardy M. Les protéines de la famille TSC-22D. Med Sci (Paris) 2015; 31:75-83. [DOI: 10.1051/medsci/20153101016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
19
|
Wong MKS, Ozaki H, Suzuki Y, Iwasaki W, Takei Y. Discovery of osmotic sensitive transcription factors in fish intestine via a transcriptomic approach. BMC Genomics 2014; 15:1134. [PMID: 25520040 PMCID: PMC4377849 DOI: 10.1186/1471-2164-15-1134] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 12/09/2014] [Indexed: 11/10/2022] Open
Abstract
Background Teleost intestine is crucial for seawater acclimation by sensing osmolality of imbibed seawater and regulating drinking and water/ion absorption. Regulatory genes for transforming intestinal function have not been identified. A transcriptomic approach was used to search for such genes in the intestine of euryhaline medaka. Results Quantitative RNA-seq by Illumina Hi-Seq Sequencing method was performed to analyze intestinal gene expression 0 h, 1 h, 3 h, 1 d, and 7 d after seawater transfer. Gene ontology (GO) enrichment results showed that cell adhesion, signal transduction, and protein phosphorylation gene categories were augmented soon after transfer, indicating a rapid reorganization of cellular components and functions. Among >50 transiently up-regulated transcription factors selected via co-expression correlation and GO selection, five transcription factors, including CEBPB and CEBPD, were confirmed by quantitative PCR to be specific to hyperosmotic stress, while others were also up-regulated after freshwater control transfer, including some well-known osmotic-stress transcription factors such as SGK1 and TSC22D3/Ostf1. Protein interaction networks suggest a high degree of overlapping among the signaling of transcription factors that respond to osmotic and general stresses, which sheds light on the interpretation of their roles during hyperosmotic stress and emergency. Conclusions Since cortisol is an important hormone for seawater acclimation as well as for general stress in teleosts, emergency and osmotic challenges could have been evolved in parallel and resulted in the overlapped signaling networks. Our results revealed important interactions among transcription factors and offer a multifactorial perspective of genes involved in seawater acclimation. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-1134) contains supplementary material, which is available to authorized users.
Collapse
|
20
|
Jäger J, Greiner V, Strzoda D, Seibert O, Niopek K, Sijmonsma TP, Schäfer M, Jones A, De Guia R, Martignoni M, Dallinga-Thie GM, Diaz MB, Hofmann TG, Herzig S. Hepatic transforming growth factor-β 1 stimulated clone-22 D1 controls systemic cholesterol metabolism. Mol Metab 2014; 3:155-66. [PMID: 24634828 DOI: 10.1016/j.molmet.2013.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 12/26/2013] [Accepted: 12/31/2013] [Indexed: 11/16/2022] Open
Abstract
Disturbances in lipid homeostasis are hallmarks of severe metabolic disorders and their long-term complications, including obesity, diabetes, and atherosclerosis. Whereas elevation of triglyceride (TG)-rich very-low-density lipoproteins (VLDL) has been identified as a risk factor for cardiovascular complications, high-density lipoprotein (HDL)-associated cholesterol confers atheroprotection under obese and/or diabetic conditions. Here we show that hepatocyte-specific deficiency of transcription factor transforming growth factor β 1-stimulated clone (TSC) 22 D1 led to a substantial reduction in HDL levels in both wild-type and obese mice, mediated through the transcriptional down-regulation of the HDL formation pathway in liver. Indeed, overexpression of TSC22D1 promoted high levels of HDL cholesterol in healthy animals, and hepatic expression of TSC22D1 was found to be aberrantly regulated in disease models of opposing energy availability. The hepatic TSC22D1 transcription factor complex may thus represent an attractive target in HDL raising strategies in obesity/diabetes-related dyslipidemia and atheroprotection.
Collapse
Affiliation(s)
- Julia Jäger
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | - Vera Greiner
- Junior Group Cellular Senescence, DKFZ, 69120 Heidelberg, Germany
| | - Daniela Strzoda
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | - Oksana Seibert
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | - Katharina Niopek
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | - Tjeerd P Sijmonsma
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | - Michaela Schäfer
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | - Allan Jones
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | - Roldan De Guia
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | - Marc Martignoni
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, Germany
| | | | - Mauricio B Diaz
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | - Thomas G Hofmann
- Junior Group Cellular Senescence, DKFZ, 69120 Heidelberg, Germany
| | - Stephan Herzig
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
21
|
Devlin RH, Sakhrani D, White S, Overturf K. Effects of domestication and growth hormone transgenesis on mRNA profiles in rainbow trout (Oncorhynchus mykiss)1. J Anim Sci 2013; 91:5247-58. [PMID: 24045478 DOI: 10.2527/jas.2013-6612] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- R. H. Devlin
- Fisheries and Oceans Canada, 4160 Marine Drive, West Vancouver, BC, Canada, V7V 1N6
| | - D. Sakhrani
- Fisheries and Oceans Canada, 4160 Marine Drive, West Vancouver, BC, Canada, V7V 1N6
| | - S. White
- Fisheries and Oceans Canada, 4160 Marine Drive, West Vancouver, BC, Canada, V7V 1N6
| | - K. Overturf
- USDA-ARS, Hagerman Fish Culture Experiment Station, 3059-F National Fish Hatchery Road, Hagerman, ID 83332
| |
Collapse
|
22
|
Riese MJ, Wang LCS, Moon EK, Joshi RP, Ranganathan A, June CH, Koretzky GA, Albelda SM. Enhanced effector responses in activated CD8+ T cells deficient in diacylglycerol kinases. Cancer Res 2013; 73:3566-77. [PMID: 23576561 PMCID: PMC3686869 DOI: 10.1158/0008-5472.can-12-3874] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Recent clinical trials have shown promise in the use of chimeric antigen receptor (CAR)-transduced T cells; however, augmentation of their activity may broaden their clinical use and improve their efficacy. We hypothesized that because CAR action requires proteins essential for T-cell receptor (TCR) signal transduction, deletion of negative regulators of these signaling pathways would enhance CAR signaling and effector T-cell function. We tested CAR activity and function in T cells that lacked one or both isoforms of diacylglycerol kinase (dgk) expressed highly in T cells, dgkα and dgkζ, enzymes that metabolize the second messenger diacylglycerol (DAG) and limit Ras/ERK activation. We found that primary murine T cells transduced with CARs specific for the human tumor antigen mesothelin showed greatly enhanced cytokine production and cytotoxicity when cocultured with a murine mesothelioma line that stably expresses mesothelin. In addition, we found that dgk-deficient CAR-transduced T cells were more effective in limiting the growth of implanted tumors, both concurrent with and after establishment of tumor. Consistent with our studies in mice, pharmacologic inhibition of dgks also augments function of primary human T cells transduced with CARs. These results suggest that deletion of negative regulators of TCR signaling enhances the activity and function of CAR-expressing T cells and identify dgks as potential targets for improving the clinical potential of CARs.
Collapse
MESH Headings
- Animals
- Blotting, Western
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/transplantation
- Cell Line, Tumor
- Cells, Cultured
- Diacylglycerol Kinase/genetics
- Diacylglycerol Kinase/immunology
- Diacylglycerol Kinase/metabolism
- Diglycerides/immunology
- Diglycerides/metabolism
- Flow Cytometry
- GPI-Linked Proteins/immunology
- HEK293 Cells
- Humans
- Immunotherapy, Adoptive
- Isoenzymes/genetics
- Isoenzymes/immunology
- Isoenzymes/metabolism
- Lymphocyte Activation/immunology
- Mesothelin
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/therapy
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Signal Transduction/immunology
Collapse
Affiliation(s)
- Matthew J. Riese
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Liang-Chuan S. Wang
- Division of Pulmonology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Edmund K. Moon
- Division of Pulmonology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Rohan P. Joshi
- Abramson Family Cancer Research Institute, Perelman School of Medicine, Philadelphia, PA
| | - Anjana Ranganathan
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Carl H. June
- Abramson Family Cancer Research Institute, Perelman School of Medicine, Philadelphia, PA
- Department of Path and Lab Medicine, Perelman School of Medicine, Philadelphia, PA
| | - Gary A. Koretzky
- Abramson Family Cancer Research Institute, Perelman School of Medicine, Philadelphia, PA
- Division of Rheumatology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Steven M. Albelda
- Division of Pulmonology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
23
|
Establishment and Validation of an Orthotopic Metastatic Mouse Model of Colorectal Cancer. ISRN HEPATOLOGY 2013; 2013:206875. [PMID: 27340651 PMCID: PMC4907346 DOI: 10.1155/2013/206875] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 03/17/2013] [Indexed: 12/24/2022]
Abstract
Metastases are largely responsible for cancer deaths in solid tumors due to the lack of effective therapies against disseminated disease, and there is an urgent need to fill this gap. This study demonstrates an orthotopic colorectal cancer (CRC) mouse model system to develop spontaneous metastasis in vivo and compare its reproducibility against human CRC. IGF1R-dependent GEO human CRC cells were used to study metastatic colonization using orthotopic transplantation procedures and demonstrated robust liver metastasis. Cell proliferation assays were performed both in the orthotopic primary colon and liver metastatic tumors, and human CRC patient's specimen and similar patterns in H&E and Ki67 staining were observed between the orthotopically generated primary and liver metastatic tumors and human CRC specimens. Microarray analysis was performed to generate gene signatures, compared with deposited human CRC gene expression data sets, analyzed by Oncomine, and revealed similarity in gene signatures with increased aggressive markers expression associated with CRC in orthotopically generated liver metastasis. Thus, we have developed an orthotopic mouse model that reproduces human CRC metastasis. This model system can be effective in developing new therapeutic strategies against disseminated disease and could be implemented for identifying genes that regulate the development and/or maintenance of established metastasis.
Collapse
|
24
|
Canterini S, Carletti V, Nusca S, Mangia F, Fiorenza MT. Multiple TSC22D4 iso-/phospho-glycoforms display idiosyncratic subcellular localizations and interacting protein partners. FEBS J 2013; 280:1320-9. [PMID: 23305244 DOI: 10.1111/febs.12123] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 12/02/2012] [Accepted: 01/01/2013] [Indexed: 12/26/2022]
Abstract
Proteins of the TSC22 domain (TSC22D) family, including TSC22D1 and TSC22D4, play pivotal roles in cell proliferation, differentiation and apoptosis, interacting with other factors in a still largely unknown manner. This study explores this issue by biochemically characterizing various TSC22D4 forms (both iso- and glyco-phospho-, namely the splice variants 42 and 55 kDa and the post-translationally modified 67 and 72 kDa forms) and their subcellular localization and protein partners during cerebellar granule neuron (CGN) differentiation. The TSC22D4-42 form is mostly cytosolic, and is the only TSC22D4 form that associates with TSC22D1.2 in undifferentiated but not differentiated CGNs. In contrast, TSC22D4-55 is prominently associated with the nuclear matrix in differentiated but not undifferentiated CGNs. As for TSC22D4-67, it is localized in the cytosol and nuclei of undifferentiated CGNs and enters mitochondria of differentiated CGNs, associating with apoptosis-inducing factor. TSC22D4-72 is modified by O-linked beta-N-acetylglucosamine (O-GlcNAcylated) and phosphorylated and is always associated with chromatin irrespective of CGN differentiation. The various subcellular localization patterns and interacting protein partners of TSC22D4 forms during CGN differentiation suggest the existence of form-specific function(s) and provide a novel framework to further investigate the biological functions of TSC22D proteins.
Collapse
Affiliation(s)
- Sonia Canterini
- Department of Psychology, Pasteur Institute-Cenci Bolognetti Foundation and Daniel Bovet Neurobiology Research Center, Sapienza University of Rome, Rome, Italy
| | | | | | | | | |
Collapse
|
25
|
Jones A, Friedrich K, Rohm M, Schäfer M, Algire C, Kulozik P, Seibert O, Müller-Decker K, Sijmonsma T, Strzoda D, Sticht C, Gretz N, Dallinga-Thie GM, Leuchs B, Kögl M, Stremmel W, Diaz MB, Herzig S. TSC22D4 is a molecular output of hepatic wasting metabolism. EMBO Mol Med 2013; 5:294-308. [PMID: 23307490 PMCID: PMC3569644 DOI: 10.1002/emmm.201201869] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 11/05/2012] [Accepted: 11/16/2012] [Indexed: 01/10/2023] Open
Abstract
In mammals, proper storage and distribution of lipids in and between tissues is essential for the maintenance of energy homeostasis. Here, we show that tumour growth triggers hepatic metabolic dysfunction as part of the cancer cachectic phenotype, particularly by reduced hepatic very-low-density-lipoprotein (VLDL) secretion and hypobetalipoproteinemia. As a molecular cachexia output pathway, hepatic levels of the transcription factor transforming growth factor beta 1-stimulated clone (TSC) 22 D4 were increased in cancer cachexia. Mimicking high cachectic levels of TSC22D4 in healthy livers led to the inhibition of hepatic VLDL release and lipogenic genes, and diminished systemic VLDL levels under both normal and high fat dietary conditions. Liver-specific ablation of TSC22D4 triggered hypertriglyceridemia through the induction of hepatic VLDL secretion. Furthermore, hepatic TSC22D4 expression levels were correlated with the degree of body weight loss and VLDL hypo-secretion in cancer cachexia, and TSC22D4 deficiency rescued tumour cell-induced metabolic dysfunction in hepatocytes. Therefore, hepatic TSC22D4 activity may represent a molecular rationale for peripheral energy deprivation in subjects with metabolic wasting diseases, including cancer cachexia.
Collapse
Affiliation(s)
- Allan Jones
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg UniversityHeidelberg, Germany
| | - Kilian Friedrich
- Dept. of Gastroenterology, University Hospital HeidelbergHeidelberg, Germany
| | - Maria Rohm
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg UniversityHeidelberg, Germany
| | - Michaela Schäfer
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg UniversityHeidelberg, Germany
| | - Carolyn Algire
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg UniversityHeidelberg, Germany
| | - Philipp Kulozik
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg UniversityHeidelberg, Germany
| | - Oksana Seibert
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg UniversityHeidelberg, Germany
| | | | - Tjeerd Sijmonsma
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg UniversityHeidelberg, Germany
| | - Daniela Strzoda
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg UniversityHeidelberg, Germany
| | - Carsten Sticht
- Medical Research Center, Klinikum MannheimMannheim, Germany
| | - Norbert Gretz
- Medical Research Center, Klinikum MannheimMannheim, Germany
| | | | | | - Manfred Kögl
- Genomics and Proteomics Core Facility, DKFZHeidelberg, Germany
| | - Wolfgang Stremmel
- Dept. of Gastroenterology, University Hospital HeidelbergHeidelberg, Germany
| | - Mauricio Berriel Diaz
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg UniversityHeidelberg, Germany
| | - Stephan Herzig
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance, Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg UniversityHeidelberg, Germany
| |
Collapse
|
26
|
Yoon CH, Rho SB, Kim ST, Kho S, Park J, Jang IS, Woo S, Kim SS, Lee JH, Lee SH. Crucial role of TSC-22 in preventing the proteasomal degradation of p53 in cervical cancer. PLoS One 2012; 7:e42006. [PMID: 22870275 PMCID: PMC3411576 DOI: 10.1371/journal.pone.0042006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 07/02/2012] [Indexed: 11/25/2022] Open
Abstract
The p53 tumor suppressor function can be compromised in many tumors by the cellular antagonist HDM2 and human papillomavirus oncogene E6 that induce p53 degradation. Restoration of p53 activity has strong therapeutic potential. Here, we identified TSC-22 as a novel p53-interacting protein and show its novel function as a positive regulator of p53. We found that TSC-22 level was significantly down-regulated in cervical cancer tissues. Moreover, over-expression of TSC-22 was sufficient to inhibit cell proliferation, promote cellular apoptosis in cervical cancer cells and suppress growth of xenograft tumors in mice. Expression of also TSC-22 enhanced the protein level of p53 by protecting it from poly-ubiquitination. When bound to the motif between amino acids 100 and 200 of p53, TSC-22 inhibited the HDM2- and E6-mediated p53 poly-ubiquitination and degradation. Consequently, ectopic over-expression of TSC-22 activated the function of p53, followed by increased expression of p21Waf1/Cip1 and PUMA in human cervical cancer cell lines. Interestingly, TSC-22 did not affect the interaction between p53 and HDM2. Knock-down of TSC-22 by small interfering RNA clearly enhanced the poly-ubiquitination of p53, leading to the degradation of p53. These results suggest that TSC-22 acts as a tumor suppressor by safeguarding p53 from poly-ubiquitination mediated-degradation.
Collapse
Affiliation(s)
- Cheol-Hee Yoon
- Division of AIDS, Center for Immunology and Pathology, National Institute of Health, Cheongwon-gun, Chungbuk, Korea
| | - Seung Bae Rho
- Research Institute, National Cancer Center, Ilsandong-gu, Goyang-si Gyeonggi-do, Korea
| | - Seong-Tae Kim
- Department of Life Science, Yongin University, Cheoin-gu, Yongin-si, Gyeonggi-do, Korea
| | - Seongho Kho
- Department of Life Science, Yongin University, Cheoin-gu, Yongin-si, Gyeonggi-do, Korea
| | - Junsoo Park
- Division of Biological Sciences and Technology, Yonsei University, Wonju, Korea
| | - Ik-Soon Jang
- Korea Basic Science Institute, Daegeon Center, Daegion, Korea
| | - Seonock Woo
- South Sea Environment Research Department, Korea Ocean Research and Development Institute, Geoje, Korea
| | - Sung Soon Kim
- Division of AIDS, Center for Immunology and Pathology, National Institute of Health, Cheongwon-gun, Chungbuk, Korea
| | - Je-Ho Lee
- School of Medicine, Sungkyunkwan University, Samsung Medical Center, Seoul, Korea
- * E-mail: (J-HL); (S-HL)
| | - Seung-Hoon Lee
- Department of Life Science, Yongin University, Cheoin-gu, Yongin-si, Gyeonggi-do, Korea
- * E-mail: (J-HL); (S-HL)
| |
Collapse
|
27
|
Sui G, Ma X, Liu S, Niu H, Dong Q. Study of the correlation between H-ras mutation and primary hepatocellular carcinoma. Oncol Lett 2012. [PMID: 23205100 DOI: 10.3892/ol.2012.832] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to investigate the correlation between H-ras mutation and primary hepatocellular carcinoma (HCC) and to describe the role of H-ras mutation in carcinogenesis. Clinical samples of 69 patients were collected and the expression levels of H-ras in HCC and the surrounding normal tissues were examined using HotStarTaq PCR. H-ras mutation was further analyzed using the PCR direct sequencing method. The results showed that H-ras mutation was present in 49 samples (49/69, 71.01%), of which 19 had codon 40 mutated from CTA to CTG and 30 had codon 61 mutated from GGC to AGC. By contrast, only 2 mutations were found in the normal tumor-adjacent tissues. The H-ras mutation rate in the high-risk of metastatic recurrence group was markedly higher than that in the low-risk group (P<0.01). The H-ras mutation rate in patients with metastatic recurrence during postoperative follow-up was also significantly higher than that in patients without metastatic recurrence (P<0.01). Based on the above results, the H-ras mutation frequency in cancer tissues is markedly higher compared with that in normal tissues. H-ras mutation may play an important role in the genesis and development of HCC and may serve as a reliable marker for individual comprehensive therapy in HCC.
Collapse
Affiliation(s)
- Guode Sui
- Departments of Emergency General Surgery and
| | | | | | | | | |
Collapse
|