1
|
Zaitseva O, Hoffmann A, Otto C, Wajant H. Targeting fibroblast growth factor (FGF)-inducible 14 (Fn14) for tumor therapy. Front Pharmacol 2022; 13:935086. [PMID: 36339601 PMCID: PMC9634131 DOI: 10.3389/fphar.2022.935086] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
Fibroblast growth factor-inducible 14 (Fn14) is a member of the tumor necrosis factor (TNF) receptor superfamily (TNFRSF) and is activated by its ligand TNF-like weak inducer of apoptosis (TWEAK). The latter occurs as a homotrimeric molecule in a soluble and a membrane-bound form. Soluble TWEAK (sTWEAK) activates the weakly inflammatory alternative NF-κB pathway and sensitizes for TNF-induced cell death while membrane TWEAK (memTWEAK) triggers additionally robust activation of the classical NF-κB pathway and various MAP kinase cascades. Fn14 expression is limited in adult organisms but becomes strongly induced in non-hematopoietic cells by a variety of growth factors, cytokines and physical stressors (e.g., hypoxia, irradiation). Since all these Fn14-inducing factors are frequently also present in the tumor microenvironment, Fn14 is regularly found to be expressed by non-hematopoietic cells of the tumor microenvironment and most solid tumor cells. In general, there are three possibilities how the tumor-Fn14 linkage could be taken into consideration for tumor therapy. First, by exploitation of the cancer associated expression of Fn14 to direct cytotoxic activities (antibody-dependent cell-mediated cytotoxicity (ADCC), cytotoxic payloads, CAR T-cells) to the tumor, second by blockade of potential protumoral activities of the TWEAK/Fn14 system, and third, by stimulation of Fn14 which not only triggers proinflammtory activities but also sensitizes cells for apoptotic and necroptotic cell death. Based on a brief description of the biology of the TWEAK/Fn14 system and Fn14 signaling, we discuss the features of the most relevant Fn14-targeting biologicals and review the preclinical data obtained with these reagents. In particular, we address problems and limitations which became evident in the preclinical studies with Fn14-targeting biologicals and debate possibilities how they could be overcome.
Collapse
Affiliation(s)
- Olena Zaitseva
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Annett Hoffmann
- Department of General, Visceral, Transplantation,Vascular and Pediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Christoph Otto
- Department of General, Visceral, Transplantation,Vascular and Pediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- *Correspondence: Harald Wajant,
| |
Collapse
|
2
|
Lower Expression of TWEAK is Associated with Poor Survival and Dysregulate TIICs in Lung Adenocarcinoma. DISEASE MARKERS 2022; 2022:8661423. [PMID: 35707713 PMCID: PMC9192298 DOI: 10.1155/2022/8661423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 02/22/2022] [Accepted: 05/03/2022] [Indexed: 12/24/2022]
Abstract
Background. Lung cancer remains the leading cause of cancer death worldwide, and the most subtype is lung adenocarcinoma (LUAD). Tumor-infiltrating immune cells (TIICs) greatly impact the prognosis of LUAD. Tumor necrosis factor–like weak inducer of apoptosis (TWEAK), signal via its receptor fibroblast growth factor-inducible 14 (Fn14), dysregulates immune cell recruitment within tumor environment, thus promoting the progression of autoimmune diseases and cancer. We aimed to explore its role in LUAD. Methods. The expression level of TWEAK was explored in Tumor Immune Estimation Resource 2.0 (TIMER2.0) and Oncomine databases. The Tumor Immune Dysfunction and Exclusion (TIDE) and Lung Cancer Explorer (LCE) databases were applied to evaluate the survival in correlation to TWEAK expression. TIICs were assessed with TIMER2.0 and TIDE datasets. The expression of TWEAK protein was detected in LUAD cell lines and also in tissue samples from LUAD patients via western blotting or combination with immunochemistry. Results. Our results showed that TWEAK was downregulated in LUAD tumors compared to normal tissues in TIMER2.0, Oncomine, cell lines, and clinical specimens. Poor survival was uncovered in lower TWEAK expression of LUAD patients in LCE (
[95% CI, 0.76-0.92]) and TCGA (
,
) and GSE13213@PRECOG (
,
) in TIDE. Multiple tumor-infiltrating immune cells (TIICs) were found closely correlated with TWEAK expression in LUAD, especially hematopoietic stem cell (
,
), common lymphoid progenitor (
,
), and myeloid-derived suppressor cells (MDSCs) (
,
). Conclusion. Lower level of TWEAK was linked with poor survival and aberrant recruitment and phenotype of TIICs in LUAD, which might motivate immune escape and weaken the effects of immunotherapy.
Collapse
|
3
|
Karimpour M, Ravanbakhsh R, Maydanchi M, Rajabi A, Azizi F, Saber A. Cancer driver gene and non-coding RNA alterations as biomarkers of brain metastasis in lung cancer: A review of the literature. Biomed Pharmacother 2021; 143:112190. [PMID: 34560543 DOI: 10.1016/j.biopha.2021.112190] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023] Open
Abstract
Brain metastasis (BM) is the most common event in patients with lung cancer. Despite multimodal treatments and advances in systemic therapies, development of BM remains one of the main factors associated with poor prognosis and mortality in patients with lung cancer. Therefore, better understanding of mechanisms involved in lung cancer brain metastasis (LCBM) is of great importance to suppress cancer cells and to improve the overall survival of patients. Several cancer-related genes such as EGFR and KRAS have been proposed as potential predictors of LCBM. In addition, there is ample evidence supporting crucial roles of non-coding RNAs (ncRNAs) in mediating LCBM. In this review, we provide comprehensive information on risk assessment, predictive, and prognostic panels for early detection of BM in patients with lung cancer. Moreover, we present an overview of LCBM molecular mechanisms, cancer driver genes, and ncRNAs which may predict the risk of BM in lung cancer patients. Recent clinical studies have focused on determining mechanisms involved in LCBM and their association with diagnosis, prognosis, and treatment outcomes. These studies have shown that alterations in EGFR, KRAS, BRAF, and ALK, as the most frequent coding gene alterations, and dysregulation of ncRNAs such as miR-423, miR-330-3p, miR-145, piR-651, and MALAT1 can be considered as potential biomarkers of LCBM.
Collapse
Affiliation(s)
- Mina Karimpour
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reyhaneh Ravanbakhsh
- Department of Aquatic Biotechnology, Artemia and Aquaculture Research Institute, Urmia University, Urmia, Iran
| | - Melika Maydanchi
- Zimagene Medical Genetics Laboratory, Avicenna St., Hamedan, Iran
| | - Ali Rajabi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Faezeh Azizi
- Genetics Office, Non-Communicable Disease Control Department, Public Health Department, Ministry of Health and Medical Education, Tehran, Iran
| | - Ali Saber
- Zimagene Medical Genetics Laboratory, Avicenna St., Hamedan, Iran.
| |
Collapse
|
4
|
Alvarez de Cienfuegos A, Cheung LH, Mohamedali KA, Whitsett TG, Winkles JA, Hittelman WN, Rosenblum MG. Therapeutic efficacy and safety of a human fusion construct targeting the TWEAK receptor Fn14 and containing a modified granzyme B. J Immunother Cancer 2020; 8:jitc-2020-001138. [PMID: 32958685 PMCID: PMC7507898 DOI: 10.1136/jitc-2020-001138] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2020] [Indexed: 12/02/2022] Open
Abstract
Background Antibody-drug conjugates are an exceptional and useful therapeutic tool for multiple diseases, particularly for cancer treatment. We previously showed that the fusion of the serine protease granzyme B (GrB), the effector molecule or T and B cells, to a binding domain allows the controlled and effective delivery of the cytotoxic payload into the target cell. The production of these constructs induced the formation of high molecular aggregates with a potential impact on the efficacy and safety of the protein. Methods Our laboratory designed a new Fn14 targeted fusion construct designated GrB(C210A)-Fc-IT4 which contains a modified GrB payload for improved protein production and preserved biological activity. We assessed the construct’s enzymatic activity, as well as in vitro cytotoxicity and internalization into target cells. We also assessed pharmacokinetics, efficacy and toxicology parameters in vivo. Results GrB(C210A)-Fc-IT4 protein exhibited high affinity and selective cytotoxicity within the nanomolar range when tested against a panel of Fn14-positive human cancer cell lines. The construct rapidly internalized into target cells, activating the caspase cascade and causing mitochondrial membrane depolarization. Pharmacokinetic studies in mice revealed that GrB(C210A)-Fc-IT4 displayed a bi-exponential clearance from plasma with a fast initial clearance (t1/2α=0.36 hour) followed by a prolonged terminal-phase plasma half-life (t1/2β=35 hours). Mice bearing MDA-MB-231 orthotopic tumor xenografts treated with vehicle or GrB(C210A)-Fc-IT4 construct (QODx5) demonstrated tumor regression and long-term (>80 days) suppression of tumor growth. Treatment of mice bearing established, subcutaneous A549 lung tumors showed impressive, long-term tumor suppression compared with a control group treated with vehicle alone. Administration of GrB(C210A)-Fc-IT4 (100 mg/kg total dose) was well-tolerated by mice and resulted in significant reduction of tumor burden in a lung cancer patient-derived xenograft model. Toxicity studies revealed no statistically significant changes in aspartate transferase, alanine transferase or lactate dehydrogenase in treated mice. Histopathological analysis of tissues from treated mice did not demonstrate any specific drug-related changes. Conclusion GrB(C210A)-Fc-IT4 demonstrated excellent, specific cytotoxicity in vitro and impressive in vivo efficacy with no significant toxicity in normal murine models. These studies show GrB(C210A)-Fc-IT4 is an excellent candidate for further preclinical development.
Collapse
Affiliation(s)
- Ana Alvarez de Cienfuegos
- Experimental Therapeutics, University of Texas MD Anderson Cancer Center Division of Cancer Medicine, Houston, Texas, USA
| | - Lawrence H Cheung
- Experimental Therapeutics, University of Texas MD Anderson Cancer Center Division of Cancer Medicine, Houston, Texas, USA
| | - Khalid A Mohamedali
- Experimental Therapeutics, University of Texas MD Anderson Cancer Center Division of Cancer Medicine, Houston, Texas, USA
| | | | - Jeffrey A Winkles
- Department of Surgery, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Walter N Hittelman
- Experimental Therapeutics, University of Texas MD Anderson Cancer Center Division of Cancer Medicine, Houston, Texas, USA
| | - Michael G Rosenblum
- Experimental Therapeutics, University of Texas MD Anderson Cancer Center Division of Cancer Medicine, Houston, Texas, USA
| |
Collapse
|
5
|
Chen Y, Cai Q, Liu S. A single molecule assay for ultrasensitive detection of Fn14 in human serum. Anal Biochem 2019; 587:113467. [PMID: 31580829 DOI: 10.1016/j.ab.2019.113467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/19/2019] [Accepted: 09/29/2019] [Indexed: 11/26/2022]
Abstract
Fibroblast growth factor inducible-14 (Fn14) is a receptor protein that plays an important role in the progression of cancer and some other diseases. Here, an ultrasensitive assay was developed for the detection of Fn14 based on a digital sandwich enzyme-linked immunosorbent bead-based assay (dELISA). Beads containing a single immunocomplex are loaded into microwells (~46 fL) and produce fluorescence through enzyme-catalyzed reactions in extremely small volumes. By measuring the number of fluorescent microwells in arrays arranged on a circular Disc, the concentration of Fn14 was determined. To obtain better performance for Fn14 detection, assay conditions including reagent concentrations and measurement parameters were optimized and 44 different antibody pairs were screened. The detection range of Fn14 is 1.26 pg/mL to 3683 pg/mL with a lower detection limit of 0.32 pg/mL, which is much lower than that of conventional ELISAs. In addition, the total operation of this assay is automated and only takes approximately an hour to accomplish. Furthermore, this assay was successfully applied to the determination of spiked Fn14 in serum samples with satisfactory performance.
Collapse
Affiliation(s)
- Yichen Chen
- Institute of Chemical Biology and Nanomedicine, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Qiyong Cai
- Institute of Chemical Biology and Nanomedicine, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| | - Song Liu
- Institute of Chemical Biology and Nanomedicine, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China.
| |
Collapse
|
6
|
Roos A, Dhruv HD, Mathews IT, Inge LJ, Tuncali S, Hartman LK, Chow D, Millard N, Yin HH, Kloss J, Loftus JC, Winkles JA, Berens ME, Tran NL. Identification of aurintricarboxylic acid as a selective inhibitor of the TWEAK-Fn14 signaling pathway in glioblastoma cells. Oncotarget 2017; 8:12234-12246. [PMID: 28103571 PMCID: PMC5355340 DOI: 10.18632/oncotarget.14685] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/26/2016] [Indexed: 12/30/2022] Open
Abstract
The survival of patients diagnosed with glioblastoma (GBM), the most deadly form of brain cancer, is compromised by the proclivity for local invasion into the surrounding normal brain, which prevents complete surgical resection and contributes to therapeutic resistance. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK), a member of the tumor necrosis factor (TNF) superfamily, can stimulate glioma cell invasion and survival via binding to fibroblast growth factor-inducible 14 (Fn14) and subsequent activation of the transcription factor NF-κB. To discover small molecule inhibitors that disrupt the TWEAK-Fn14 signaling axis, we utilized a cell-based drug-screening assay using HEK293 cells engineered to express both Fn14 and a NF-κB-driven firefly luciferase reporter protein. Focusing on the LOPAC1280 library of 1280 pharmacologically active compounds, we identified aurintricarboxylic acid (ATA) as an agent that suppressed TWEAK-Fn14-NF-κB dependent signaling, but not TNFα-TNFR-NF-κB driven signaling. We demonstrated that ATA repressed TWEAK-induced glioma cell chemotactic migration and invasion via inhibition of Rac1 activation but had no effect on cell viability or Fn14 expression. In addition, ATA treatment enhanced glioma cell sensitivity to both the chemotherapeutic agent temozolomide (TMZ) and radiation-induced cell death. In summary, this work reports a repurposed use of a small molecule inhibitor that targets the TWEAK-Fn14 signaling axis, which could potentially be developed as a new therapeutic agent for treatment of GBM patients.
Collapse
Affiliation(s)
- Alison Roos
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, Arizona 85259, USA
| | - Harshil D Dhruv
- Cancer and Cell Biology Division, The Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Ian T Mathews
- Cancer and Cell Biology Division, The Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Landon J Inge
- Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, AZ 85004, USA
| | - Serdar Tuncali
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, Arizona 85259, USA
| | - Lauren K Hartman
- Cancer and Cell Biology Division, The Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Donald Chow
- Cancer and Cell Biology Division, The Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Nghia Millard
- Cancer and Cell Biology Division, The Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Holly H Yin
- Cancer and Cell Biology Division, The Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Jean Kloss
- Department of Biochemistry and Molecular Biology, Mayo Clinic Arizona, Scottsdale, Arizona 85259, USA
| | - Joseph C Loftus
- Department of Biochemistry and Molecular Biology, Mayo Clinic Arizona, Scottsdale, Arizona 85259, USA
| | - Jeffrey A Winkles
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Michael E Berens
- Cancer and Cell Biology Division, The Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Nhan L Tran
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, Arizona 85259, USA
| |
Collapse
|
7
|
Affiliation(s)
- Guanglei Hu
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an, China
| | - Weihui Zeng
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
8
|
Yu R, Han L, Ni X, Wang M, Xue P, Zhang L, Yuan M. Kruppel-like factor 4 inhibits non–small cell lung cancer cell growth and aggressiveness by stimulating transforming growth factor-β1-meidated ERK/JNK/NF-κB signaling pathways. Tumour Biol 2017. [PMID: 28631556 DOI: 10.1177/1010428317705574] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Affiliation(s)
- Renzhi Yu
- Department of Respiratory Medicine, Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, China
| | - Lei Han
- Department of Respiratory Medicine, Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, China
| | - Xin Ni
- Department of Respiratory Medicine, Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, China
| | - Minghuan Wang
- Community Health Service Center, Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, China
| | - Ping Xue
- Department of Respiratory Medicine, Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, China
| | - Li Zhang
- Department of Respiratory Medicine, Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, China
| | - Mei Yuan
- Department of Respiratory Medicine, Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, China
| |
Collapse
|
9
|
Sequera C, Vázquez-Carballo A, Arechederra M, Fernández-Veledo S, Porras A. TWEAK promotes migration and invasion in MEFs through a mechanism dependent on ERKs activation and Fibulin 3 down-regulation. J Cell Physiol 2017; 233:968-978. [PMID: 28383766 DOI: 10.1002/jcp.25942] [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: 07/05/2016] [Accepted: 03/30/2017] [Indexed: 11/10/2022]
Abstract
TWEAK regulates multiple physio-pathological processes in fibroblasts such as fibrosis. It also induces migration and invasion in tumors and it can activate p38 MAPK in various cell types. Moreover, p38α MAPK promotes migration and invasion in several cancer cells types and in mouse embryonic fibroblasts (MEFs). However, it remains unknown if TWEAK could promote migration in fibroblasts and whether p38α MAPK might play a role. Our results reveal that TWEAK activates ERKs, Akt, and p38α/β MAPKs and reduces secreted Fibulin 3 in MEFs. TWEAK also increases migration and invasion in wt and p38α deficient MEFs, which indicates that p38α MAPK is not required to mediate these effects. In contrast, ERKs inhibition significantly decreases TWEAK-induced migration and Fibulin 3 knock-down mimics TWEAK effect. These results indicate that both ERKs activation and Fibulin 3 down-regulation would contribute to mediate TWEAK pro-migratory effect. In fact, the additional regulation of ERKs and/or p38β as a consequence of Fibulin 3 decrease might be also involved in the pro-migratory effect of TWEAK in MEFs. In conclusion, our studies uncover novel mechanisms by which TWEAK would favor tissue repair by promoting fibroblasts migration.
Collapse
Affiliation(s)
- Celia Sequera
- Facultad de Farmacia, Departamento de Bioquímica y Biología Molecular II, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Ana Vázquez-Carballo
- Facultad de Farmacia, Departamento de Bioquímica y Biología Molecular II, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - María Arechederra
- Facultad de Farmacia, Departamento de Bioquímica y Biología Molecular II, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Sonia Fernández-Veledo
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.,Hospital Universitari de Tarragona Joan XXIII, IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Almudena Porras
- Facultad de Farmacia, Departamento de Bioquímica y Biología Molecular II, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| |
Collapse
|
10
|
Walker SJ, Beavers DP, Fortunato J, Krigsman A. A Putative Blood-Based Biomarker for Autism Spectrum Disorder-Associated Ileocolitis. Sci Rep 2016; 6:35820. [PMID: 27767057 PMCID: PMC5073317 DOI: 10.1038/srep35820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/06/2016] [Indexed: 12/11/2022] Open
Abstract
Gastrointestinal symptoms are common in children with autism spectrum disorder (ASD). A significant proportion of children with ASD and gastrointestinal symptoms have histologic evidence of ileocolitis (inflammation of the terminal ileum and/or colon). We previously reported the molecular characterization of gastrointestinal biopsy tissue from ASD children with ileocolitis (ASDIC+) compared to anatomically similar inflamed tissue from typically developing children with inflammatory bowel disease (IBD; i.e. Crohn’s disease or ulcerative colitis) and typically developing children with gastrointestinal symptoms but no evidence of gastrointestinal mucosal inflammation (TDIC−). ASDIC+ children had a gene expression profile that, while primarily overlapping with known IBD, had distinctive differences. The present study confirms these findings and replicates this molecular characterization in a second cohort of cases (ASDIC+) and controls (TDIC−). In these two separate case/control mucosal-based cohorts, we have demonstrated overlap of 59 differentially expressed transcripts (DETs) unique to inflamed ileocolonic tissue from symptomatic ASDIC+ children. We now report that 9 of these 59 transcripts are also differentially expressed in the peripheral blood of the second cohort of ASDIC+ children. This set of transcripts represents a putative blood-based biomarker for ASD-associated ileocolonic inflammation.
Collapse
Affiliation(s)
- Stephen J Walker
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston Salem, NC, USA
| | - Daniel P Beavers
- Department of Biostatistical Sciences, Public Health Sciences, Wake Forest University Health Sciences, Winston Salem, NC, USA
| | - John Fortunato
- Pediatric Gastroenterology, Hepatology, and Nutrition, Ann &Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Arthur Krigsman
- Pediatric Gastroenterology Resources, 148 Beach 9th Street, Suite 2B, Far Rockaway, NY, USA
| |
Collapse
|
11
|
Wang H, Wang Y, Du Q, Lu P, Fan H, Lu J, Hu R. Inflammasome-independent NLRP3 is required for epithelial-mesenchymal transition in colon cancer cells. Exp Cell Res 2016; 342:184-92. [PMID: 26968633 DOI: 10.1016/j.yexcr.2016.03.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/07/2016] [Accepted: 03/07/2016] [Indexed: 12/22/2022]
Abstract
Inflammasome NLRP3 plays a crucial role in the process of colitis and colitis--associated colon cancer. Even though much is known regarding the NLRP3 inflammasome that regulates pro-inflammatory cytokine release in innate immune cells, the role of NLRP3 in non-immune cells is still unclear. In this study, we showed that NLRP3 was highly expressed in mesenchymal-like colon cancer cells (SW620), and was upregulated by tumor necrosis factors-α (TNF-α) and transforming growth factor-β1 (TGF-β1) respectively, during EMT in colon cancer epithelial cells HCT116 and HT29. Knockdown of NLRP3 retained epithelial spindle-like morphology of HCT116 and HT29 cells and reversed the mesenchymal characteristic of SW620 cells, indicated by the decreased expression of vimentin and MMP9 and increased expression of E-cadherin. In addition, knockdown of NLRP3 in colorectal carcinoma cells displayed diminished cell migration and invasion. Interestingly, during the EMT process induced by TNF-α or TGF-β1, the cleaved caspase-1 and ASC speck were not detected, indicating that NLRP3 functions in an inflammasome-independent way. Further studies demonstrated that NLRP3 protein expression was regulated by NF-κB signaling in TNF-α or TGF-β1-induced EMT, as verified by the NF-κB inhibitor Bay 11-7082. Moreover, NLRP3 knockdown reduced the expression of Snail1, indicating that NLRP3 may promote EMT through regulating Snail1. In summary, our results showed that the NLRP3 expression, not the inflammasome activation, was required for EMT in colorectal cancer cells.
Collapse
Affiliation(s)
- Hong Wang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Yajing Wang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Qianming Du
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Ping Lu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Huimin Fan
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Jinrong Lu
- Department of Organic Chemistry, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Rong Hu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
12
|
Meulendijks D, Lassen UN, Siu LL, Huitema ADR, Karanikas V, Mau-Sorensen M, Jonker DJ, Hansen AR, Simcox ME, Schostack KJ, Bottino D, Zhong H, Roessler M, Vega-Harring SM, Jarutat T, Geho D, Wang K, DeMario M, Goss GD, Schellens JHM. Exposure and Tumor Fn14 Expression as Determinants of Pharmacodynamics of the Anti-TWEAK Monoclonal Antibody RG7212 in Patients with Fn14-Positive Solid Tumors. Clin Cancer Res 2015; 22:858-67. [PMID: 26446946 DOI: 10.1158/1078-0432.ccr-15-1506] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/12/2015] [Indexed: 12/16/2022]
Abstract
PURPOSE The TWEAK-Fn14 pathway represents a novel anticancer target that is being actively investigated. Understanding the relationship between pharmacokinetics of anti-TWEAK therapeutics and tumor pharmacodynamics is critical. We investigated exposure-response relationships of RG7212, an anti-TWEAK mAb, in patients with Fn14-expressing tumors. EXPERIMENTAL DESIGN Patients with Fn14-positive tumors (IHC ≥ 1+) treated in a phase I first-in-human study with ascending doses of RG7212 were the basis for this analysis. Pharmacokinetics of RG7212 and dynamics of TWEAK were determined, as were changes in tumor TWEAK-Fn14 signaling in paired pre- and posttreatment tumor biopsies. The objectives of the analysis were to define exposure-response relationships and the relationship between pretreatment tumor Fn14 expression and pharmacodynamic effect. Associations between changes in TWEAK-Fn14 signaling and clinical outcome were explored. RESULTS Thirty-six patients were included in the analysis. RG7212 reduced plasma TWEAK to undetectable levels at all observed RG7212 exposures. In contrast, reductions in tumor Fn14 and TRAF1 protein expression were observed only at higher exposure (≥ 300 mg*h/mL). Significant reductions in tumor Ki-67 expression and early changes in serum concentrations of CCL-2 and MMP-9 were observed exclusively in patients with higher drug exposure who had high pretreatment tumor Fn14 expression. Pretreatment tumor Fn14 expression was not associated with outcome, but a trend toward longer time on study was observed with high versus low RG7212 exposure. CONCLUSIONS RG7212 reduced tumor TWEAK-Fn14 signaling in a systemic exposure-dependent manner. In addition to higher exposure, relatively high Fn14 expression might be required for pharmacodynamic effect of anti-TWEAK monoclonal antibodies.
Collapse
Affiliation(s)
- Didier Meulendijks
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ulrik N Lassen
- Department of Oncology, The Finsen Centre, Rigshospitalet, Copenhagen, Denmark
| | - Lillian L Siu
- Ontario Cancer Institute/Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Vaios Karanikas
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Morten Mau-Sorensen
- Department of Oncology, The Finsen Centre, Rigshospitalet, Copenhagen, Denmark
| | | | - Aaron R Hansen
- Ontario Cancer Institute/Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Mary E Simcox
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, New York, New York
| | - Kathleen J Schostack
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, New York, New York
| | - Dean Bottino
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, New York, New York
| | - Hua Zhong
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, New York, New York
| | - Markus Roessler
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Penzberg, Germany
| | - Suzana M Vega-Harring
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Penzberg, Germany
| | - Tiantom Jarutat
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Penzberg, Germany
| | - David Geho
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, New York, New York
| | - Karen Wang
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, New York, New York
| | - Mark DeMario
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, New York, New York
| | | | - Jan H M Schellens
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands. Department of Pharmaceutical Sciences, Science Faculty, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
13
|
The TWEAK receptor Fn14 is a potential cell surface portal for targeted delivery of glioblastoma therapeutics. Oncogene 2015; 35:2145-55. [PMID: 26300004 DOI: 10.1038/onc.2015.310] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/14/2015] [Accepted: 07/14/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Fibroblast growth factor-inducible 14 (Fn14; TNFRSF12A) is the cell surface receptor for the tumor necrosis factor (TNF) family member TNF-like weak inducer of apoptosis (TWEAK). The Fn14 gene is normally expressed at low levels in healthy tissues but expression is significantly increased after tissue injury and in many solid tumor types, including glioblastoma (GB; formerly referred to as 'GB multiforme'). GB is the most common and aggressive primary malignant brain tumor and the current standard-of-care therapeutic regimen has a relatively small impact on patient survival, primarily because glioma cells have an inherent propensity to invade into normal brain parenchyma, which invariably leads to tumor recurrence and patient death. Despite major, concerted efforts to find new treatments, a new GB therapeutic that improves survival has not been introduced since 2005. In this review article, we summarize studies indicating that (i) Fn14 gene expression is low in normal brain tissue but is upregulated in advanced brain cancers and, in particular, in GB tumors exhibiting the mesenchymal molecular subtype; (ii) Fn14 expression can be detected in glioma cells residing in both the tumor core and invasive rim regions, with the maximal levels found in the invading glioma cells located within normal brain tissue; and (iii) TWEAK Fn14 engagement as well as Fn14 overexpression can stimulate glioma cell migration, invasion and resistance to chemotherapeutic agents in vitro. We also discuss two new therapeutic platforms that are currently in development that leverage Fn14 overexpression in GB tumors as a way to deliver cytotoxic agents to the glioma cells remaining after surgical resection while sparing normal healthy brain cells.
Collapse
|
14
|
Wang J, Yan B, Liu SM, Sun H, Pan Y, Guan D, Zhang X, Xu J, Ma H. Transcriptomic and Functional Pathway Analysis of Human Cervical Carcinoma Cancer Cells Response to Microtubule Inhibitor. J Cancer 2015; 6:930-937. [PMID: 26316889 PMCID: PMC4543753 DOI: 10.7150/jca.12284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/12/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND There clearly is a need for effective chemotherapy for early-stage, high-risk patients with human cervical carcinoma. Vinblastine (VBL) is a key microtubule inhibitor, but unproven in its mechanisms as an important antitumor agent in cervical carcinoma. METHODS We selected the concentration of vinblastine inducing 30% cell death for analyses assessing the DNA content, gene expression and transcriptional gene regulation of VBL-treated KB-3 cells. RESULTS Transcriptomic and hierarchical clustering analysis demonstrated that treatment of KB-3 cells with VBL altered the expression of a diverse group of genes with G2/M arrest, which regulated by four oncogenic or tumor suppresser transcription factors (AP1, NFKB1, RELA, and TP53). Functional pathway analysis revealed the disease response to the biological effects of vinblastine in cervical carcinoma chemotherapy including protein ubiquitination pathway, RhoGDI signaling, integrin signaling, agranulocyte adhesion and biapedesis, and actin nucleation pathways. Northern blots also confirmed that KRT-7, FN14, IER3, and ID1 were deregulated in VBL-treated KB-3 cells. CONCLUSION Transcriptional time series profiles and a functional pathway analysis of VBL-treated KB-3 cells will provide a new strategy for improving microtubule inhibitor chemotherapy for cervical carcinoma.
Collapse
Affiliation(s)
- Jin Wang
- 1. Scientific Research Center, Shanghai Public Health Clinical Center, 2901 Caolang Road, Jinshan District, Shanghai 201508, China
- 2. Department of Translational Molecular Pathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Bin Yan
- 3. Laboratory for Food Safety and Environmental Technology, Institutes of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Song-Mei Liu
- 4. Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Huanhuan Sun
- 5. Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Yonglong Pan
- 3. Laboratory for Food Safety and Environmental Technology, Institutes of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Daogang Guan
- 6. Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Xiaoyan Zhang
- 1. Scientific Research Center, Shanghai Public Health Clinical Center, 2901 Caolang Road, Jinshan District, Shanghai 201508, China
| | - Jianqing Xu
- 1. Scientific Research Center, Shanghai Public Health Clinical Center, 2901 Caolang Road, Jinshan District, Shanghai 201508, China
| | - Haiqing Ma
- 5. Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| |
Collapse
|
15
|
A high-fat diet is associated with altered adipokine production and a more aggressive esophageal adenocarcinoma phenotype in vivo. J Thorac Cardiovasc Surg 2014; 149:1185-91. [PMID: 25595377 DOI: 10.1016/j.jtcvs.2014.11.076] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 11/18/2014] [Accepted: 11/28/2014] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Obesity has been linked to esophageal adenocarcinoma (EAC). We hypothesize that adipokines, which are altered by obesity, could affect EAC growth rates and potentially serve as biomarkers of disease and targets for treatment. We have developed a potential murine model to investigate the effects of obesity-altered adipokines on EAC in vivo. METHODS Severe combined immune-deficient mice were fed either a high-fat diet (HFD) containing 60% animal fat, or a control diet with 10% animal fat, and monitored for weight gain for 5 weeks. All mice were subcutaneously implanted with EAC cells (OE33), and tumor volume was monitored for an additional 4 weeks by direct measurement and uptake of fluorescently labeled 2-D-deoxyglucose. At sacrifice, serum triglyceride levels and abdominal fat-pad weight were measured to assess obesity state. Adipokine levels were measured within abdominal fat of tumor-bearing mice. RESULTS Mice fed the HFD displayed increased body weight, visceral fat, and serum leptin and triglycerides. All mice developed tumors; OE33 EAC cells in HFD mice displayed increased growth rates, proliferation, and metabolic activity relative to tumors of EAC in control diet mice. Adipokine expression in the abdominal fat revealed distinct changes associated with the HFD and increased body weight. CONCLUSIONS Ad libitum feeding of the HFD was correlated with more-proliferative EAC tumors in vivo. This phenotype was associated with alterations to secreted adipokines, representing a potential mechanism for our observations. Further studies are necessary to explore findings, as they have potential to improve treatment of EAC.
Collapse
|
16
|
Cheng E, Whitsett TG, Tran NL, Winkles JA. The TWEAK Receptor Fn14 Is an Src-Inducible Protein and a Positive Regulator of Src-Driven Cell Invasion. Mol Cancer Res 2014; 13:575-83. [PMID: 25392346 DOI: 10.1158/1541-7786.mcr-14-0411] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED The TNF receptor superfamily member Fn14 (TNFRSF12A) is the sole signaling receptor for the proinflammatory cytokine TWEAK (TNFSF12). TWEAK Fn14 engagement stimulates multiple signal transduction pathways, including the NF-κB pathway, and this triggers important cellular processes (e.g., growth, differentiation, migration, and invasion). The TWEAK-Fn14 axis is thought to be a major physiologic mediator of tissue repair after acute injury. Various studies have revealed that Fn14 is highly expressed in many solid tumor types, and that Fn14 signaling may play a role in tumor growth and metastasis. Previously, it was shown that Fn14 levels are frequently elevated in non-small cell lung cancer (NSCLC) tumors and cell lines that exhibit constitutive EGFR phosphorylation (activation). Furthermore, elevated Fn14 levels increased NSCLC cell invasion in vitro and lung metastatic tumor colonization in vivo. The present study reveals that EGFR-mutant NSCLC cells that express high levels of Fn14 exhibit constitutive activation of the cytoplasmic tyrosine kinase Src, and that treatment with the Src family kinase (SFK) inhibitor dasatinib decreases Fn14 gene expression at both the mRNA and protein levels. Importantly, siRNA-mediated depletion of the SFK member Src in NSCLC cells also decreases Fn14 expression. Finally, expression of the constitutively active v-Src oncoprotein in NIH 3T3 cells induces Fn14 gene expression, and NIH 3T3/v-Src cells require Fn14 expression for full invasive capacity. IMPLICATIONS These results indicate that oncogenic Src may contribute to Fn14 overexpression in solid tumors, and that Src-mediated cell invasion could potentially be inhibited with Fn14-targeted therapeutics.
Collapse
Affiliation(s)
- Emily Cheng
- Department of Surgery, Center for Vascular and Inflammatory Diseases, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Timothy G Whitsett
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Nhan L Tran
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Jeffrey A Winkles
- Department of Surgery, Center for Vascular and Inflammatory Diseases, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|