1
|
Suresh D, Mukherjee S, Zambre A, Ghoshdastidar S, Yadavilli S, Rekha KR, Upendran A, Kannan R. Nanoparticle-Mediated Cosilencing of Drug Resistance and Compensatory Genes Enhances Lung Cancer Therapy. ACS NANO 2025; 19:15256-15271. [PMID: 40239042 DOI: 10.1021/acsnano.4c12318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Non-small cell lung cancer (NSCLC) is challenging to treat due to acquired drug resistance, leading to high mortality rates. NSCLC patients with mutations in the epidermal growth factor receptor (EGFR) region are treated with tyrosine kinase inhibitors (TKI) as a first-line treatment, but many develop resistance within 1-2 years. AXL overexpression contributes to drug resistance in over 25% of patients, as shown by tumor analyses, prompting efforts to develop small-molecule inhibitors targeting AXL. However, we found that AXL repression increases compensatory FN14 signaling that could affect the therapeutic efficacy. Therefore, we chose to evaluate therapeutic efficacy after silencing both AXL and FN14 genes using short interfering RNA (siRNA) therapy. While siRNAs are more selective than small-molecule inhibitors, they are prone to in vivo degradation. To address this, we developed gelatin nanoparticles carrying siRNAs targeting AXL and FN14 (GsiAF). These nanoparticles were designed to protect siRNA from serum degradation and to allow antibody functionalization on their surface. We demonstrate that GsiAF selectively and effectively silences the respective genes under both in vitro and in vivo conditions, thereby overcoming compensatory FN14 signaling. Results indicate that GsiAF was successful in delivering siRNAs to tumors and downregulating both AXL and FN14 genes. We show that coinhibition of AXL and FN14 has effectively decreased TKI resistance in cancer cells and significantly reduced tumor growth in mice bearing lung cancer. The gelatin-siRNA nanoconstruct combined with TKI represents a promising strategy for overcoming drug resistance in NSCLC and other cancers, with potential for future clinical translation.
Collapse
MESH Headings
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Humans
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Animals
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
- Receptor Protein-Tyrosine Kinases/metabolism
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Nanoparticles/chemistry
- Mice
- Axl Receptor Tyrosine Kinase
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/chemistry
- Cell Line, Tumor
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/chemistry
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Mice, Nude
Collapse
Affiliation(s)
- Dhananjay Suresh
- Department of Bioengineering, University of Missouri, Columbia, Missouri 65212, United States
- Department of Radiology, University of Missouri, Columbia, Missouri 65212, United States
| | - Soumavo Mukherjee
- Department of Bioengineering, University of Missouri, Columbia, Missouri 65212, United States
| | - Ajit Zambre
- Department of Radiology, University of Missouri, Columbia, Missouri 65212, United States
| | - Shreya Ghoshdastidar
- Department of Bioengineering, University of Missouri, Columbia, Missouri 65212, United States
| | - Sairam Yadavilli
- Department of Bioengineering, University of Missouri, Columbia, Missouri 65212, United States
| | - Karamkolly R Rekha
- Department of Radiology, University of Missouri, Columbia, Missouri 65212, United States
| | - Anandhi Upendran
- Department of Medical Pharmacology & Physiology, University of Missouri, Columbia, Missouri 65212, United States
| | - Raghuraman Kannan
- Department of Bioengineering, University of Missouri, Columbia, Missouri 65212, United States
- Department of Radiology, University of Missouri, Columbia, Missouri 65212, United States
- Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, Missouri 65212, United States
| |
Collapse
|
2
|
Fu W, Liang Q, Ma Y, Lei S, Li R, Zheng X, Chen L, Chen J, Cai X, Dai X, Duan H, He W, Ren J. Fn14-targeting, NIR-II responsive nanomaterials for enhanced radiotherapy against glioblastomas. NANOSCALE ADVANCES 2025; 7:2634-2647. [PMID: 40109505 PMCID: PMC11915457 DOI: 10.1039/d4na00788c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/01/2025] [Indexed: 03/22/2025]
Abstract
Radiotherapy is a common treatment option for patients with glioblastoma multiforme. However, tumor heterogeneity causes varying responses to radiation among different tumor subpopulations. Cancer cells that endure radiotherapy exhibit radioresistance, resulting in the ineffectiveness of radiation therapy and eventual tumor relapse. In this study, we discovered that the fibroblast growth factor-inducible 14 (Fn14)-positive tumor cells were enriched in tumor residual foci after radiation, ultimately leading to treatment failure. Fn14-expressing glioma cells survived ionizing radiation through preferential activation of DNA damage checkpoint response. We have thus engineered an Fn14-targeting and NIR-II responsive plasmonic gold nanosystem named Fn14-AuNPs, which can precisely internalize into Fn14-overexpressed glioma cells and have an excellent BBB-crossing capability. As gold nanoparticles, by inhibition of DNA repair processes and induction of G2/M cells cycle arrest, Fn14-AuNPs nanoparticles improved the radiosensitivity of tumor cells. Meanwhile, Fn14-AuNPs induced localized heat under NIR-II photoirradiation, thus impeding RT-induced DNA damage checkpoint response. This versatile nanosensitizer, combined with NIR-II laser photoirradiation, can eradicate radioresistant subpopulations of glioblastoma and improve the therapeutic effect of radiotherapy. This finding presents an effective radiosensitization strategy by targeting radioresistant subpopulations, which can efficiently overcome the constraints imposed in clinical radiotherapy and offer a hopeful avenue to enhance the treatment effectivity of radiotherapy in glioblastoma.
Collapse
Affiliation(s)
- Wei Fu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Qing Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Yuxi Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Shiqiong Lei
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
| | - Ruiqi Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Xin Zheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Lian Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Jiayuan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Xing Cai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Xiaofang Dai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
| | - Hongwei Duan
- School of Chemical and Biomedical Engineering, Nanyang Technological University Singapore 637457 Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore 636921 Singapore
| | - Wenshan He
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
| | - Jinghua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Hubei Key Laboratory of Precision Radiation Oncology Wuhan 430022 China
- Shenzhen Huazhong University of Science and Technology Research Institute Shenzhen 518057 China
| |
Collapse
|
3
|
Oblitas CM, Sampedro-Viana A, Fernández-Rodicio S, Rodríguez-Yáñez M, López-Dequidt I, Gonzalez-Quintela A, Mosqueira AJ, Porto-Álvarez J, Fernández JM, Bazarra-Barreiros M, Abengoza-Bello MT, Ortega-Espina S, Ouro A, Campos F, Sobrino T, Castillo J, Alonso-Alonso ML, Hervella P, Iglesias-Rey R. Hyperthermia and Early Growth of Cerebral Infarct: The Potential Role of Blood-Brain Barrier Permeability. Transl Stroke Res 2025:10.1007/s12975-025-01349-x. [PMID: 40195239 DOI: 10.1007/s12975-025-01349-x] [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: 01/20/2025] [Revised: 03/10/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
Hyperthermia within the first 24 h following ischemic stroke (IS) has been associated with poor outcomes. We sought to determine whether blood-brain barrier (BBB) permeability contributes to the relationship between hyperthermia and early infarct growth (EIG). A retrospective analysis was conducted on a prospective stroke biobank. EIG was defined as the percentage difference between the initial volume (mL) determined by the diffusion-weighted imaging at admission and the volume (mL) from the control CT image on the 4 th-7 th day. Hyperthermia was defined as an axillary body temperature ≥ 37.5 °C within the first 24 h. Soluble tumor necrosis factor-like weak inducer of apoptosis (sTWEAK) serum levels were measured by ELISA. One-hundred and two (19.7%) patients showed EIG from a cohort of 519 patients (45.6% females). Linear correlation was observed for axillar body temperature and EIG (Pearson's r = 0.46; p < 0.001). sTWEAK serum levels showed a c-statistic of 0.74 (95% CI: 0.69-0.79), with an optimal cut-off point > 3000 pg/mL for EIG prediction. Moreover, microalbuminuria levels strongly correlated with sTWEAK levels (Pearson's r = 0.75; p < 0.001). In the multivariate analysis for EIG was observed an independent association with hyperthermia (adjusted OR 24.21; 95% CI: 12.03-39.12), sTWEAK levels > 3000 pg/mL (adjusted OR 16.43; 95% CI: 3.71-72.70), leukoaraiosis (adjusted OR 10.42; 95% CI: 2.68-39.08), and microalbuminuria (adjusted OR 1.02; 95% CI: 1.00-1.12). In our cohort, hyperthermia was independently associated with EIG after IS. The fact that microalbuminuria, leukoaraiosis, and sTWEAK were also associated with EIG suggests a relationship with increased BBB permeability.
Collapse
Grants
- CM23/00173 Instituto de Salud Carlos III
- FI22/00200 Instituto de Salud Carlos III
- CP14/00154 Instituto de Salud Carlos III
- CPII17/00027 Instituto de Salud Carlos III
- PI17/01103, PI22/00938, ISCIII/PI21/01256 Instituto de Salud Carlos III
- PI17/01103, PI22/00938, ISCIII/PI21/01256 Instituto de Salud Carlos III
- PI17/01103, PI22/00938, ISCIII/PI21/01256 Instituto de Salud Carlos III
- PI17/01103, PI22/00938, ISCIII/PI21/01256 Instituto de Salud Carlos III
- CB22/05/00067 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas
- CB22/05/00067 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas
- CB22/05/00067 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas
- CB22/05/00067 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas
- CB22/05/00067 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas
- CB22/05/00067 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas
- SAF2017-84267-R Ministerio de Ciencia e Innovación
- SAF2017-84267-R Ministerio de Ciencia e Innovación
- SAF2017-84267-R Ministerio de Ciencia e Innovación
- SAF2017-84267-R Ministerio de Ciencia e Innovación
- IN607A2022-03, IN607A2022/07 Xunta de Galicia
- IN607A2022-03, IN607A2022/07 Xunta de Galicia
- IN607A2022-03, IN607A2022/07 Xunta de Galicia
- IN607A2022-03, IN607A2022/07 Xunta de Galicia
- 0624_2IQBIONEURO_6_E POCTEP
- 0624_2IQBIONEURO_6_E POCTEP
- 0624_2IQBIONEURO_6_E POCTEP
- 0624_2IQBIONEURO_6_E POCTEP
Collapse
Affiliation(s)
- Crhistian-Mario Oblitas
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain.
| | - Ana Sampedro-Viana
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
| | - Sabela Fernández-Rodicio
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
| | - Manuel Rodríguez-Yáñez
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, Santiago de Compostela, 15706, A Coruña, Spain
| | - Iria López-Dequidt
- Department of Neurology, Hospital Clínico Universitario de Ferrol, Ferrol, 15405, A Coruña, Spain
| | - Arturo Gonzalez-Quintela
- Department of Internal Medicine, Hospital Clínico Universitario, Santiago de Compostela, 15706, A Coruña, Spain
| | - Antonio J Mosqueira
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
- Department of Neuroradiology, Hospital Clínico Universitario, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
| | - Jacobo Porto-Álvarez
- Department of Neuroradiology, Hospital Clínico Universitario, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
| | - Javier Martínez Fernández
- Department of Neuroradiology, Hospital Clínico Universitario, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
| | - Marcos Bazarra-Barreiros
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
| | - María Teresa Abengoza-Bello
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
| | - Sara Ortega-Espina
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
| | - Alberto Ouro
- Neuroaging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Francisco Campos
- Translational Stroke Laboratory (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
| | - Tomás Sobrino
- Neuroaging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
| | - Maria Luz Alonso-Alonso
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
| | - Pablo Hervella
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, A Coruña, Spain.
| |
Collapse
|
4
|
Vasilijic S, Seist R, Yin Z, Xu L, Stankovic KM. Immune profiling of human vestibular schwannoma secretions identifies TNF-α and TWEAK as cytokines with synergistic potential to impair hearing. J Neuroinflammation 2025; 22:35. [PMID: 39923035 PMCID: PMC11807327 DOI: 10.1186/s12974-025-03364-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/30/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Vestibular schwannoma (VS) is an intracranial tumor arising from the Schwann cells of the vestibular nerve and is an important cause of sensorineural hearing loss (SNHL) in humans. The mechanisms underlying this SNHL are incompletely understood and currently, there are no drugs FDA approved specifically for VS. This knowledge gap significantly limits the development of effective treatments aimed at preventing, stabilizing, or reversing VS-induced SNHL. METHODS To identify effector molecules involved in VS-induced SNHL, we analyzed 47 immune-related factors secreted by tumor tissue in over 50 patients with sporadic VS and studied their correlation with preoperative hearing ability and tumor size. The most promising effector molecules were validated in vivo in an anatomically accurate mouse model of VS, and in vitro with mouse fibroblasts (L929) and auditory cell lines representing pro-sensory precursors of hair cells (UB-OC1) and auditory neuroblasts (US-VOT-N33). RESULTS We demonstrated that VS-induced SNHL was linked to increased secretion of TNF-α, IL-2R, CD163, eotaxin, and HGF, while larger tumor size was associated with higher levels of TNF-α, TNF-R2, IL-1α, IFN-α, MIP-1β, and IL-21 secretion. We identified heterogeneity among VS tumors in their capacity to secrete TNF-α. Tumors with high levels of TNF-α secretion released cytokines and chemokines that significantly correlated with poor hearing (TWEAK and eotaxin) or better hearing (LIF, GRO-α, MIP-1α, MIP-3α, and IL-1α). Among these, TWEAK was notably abundant, with levels exceeding those in normal nerve tissue, elevated in patients with non-serviceable hearing and strongly linked to poor hearing in patients with TNF-α high-secreting tumors. In vivo, we demonstrated that VS-secreted factors reach the inner ear, with elevated TNF-α and TWEAK in the perilymph and blood of tumor-bearing mice with impaired hearing. In vitro, TWEAK amplified TNF-α -mediated cytotoxicity in TNF-α sensitive cells (L929) and auditory cell lines (UB-OC1 and US-VOT-N33) at tumor-secreted concentrations. CONCLUSION This study provides compelling evidence that VS-secreted TNF-α and TWEAK act synergistically to drive tumor-induced SNHL. Targeting the TNF-α/TWEAK axis presents a promising new avenue for preventing VS-induced SNHL.
Collapse
Affiliation(s)
- Sasa Vasilijic
- Department of Otolaryngology- Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Rd, Palo Alto, Stanford, CA, 94304, USA
| | - Richard Seist
- Department of Otolaryngology- Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Rd, Palo Alto, Stanford, CA, 94304, USA
| | - Zhenzhen Yin
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lei Xu
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Konstantina M Stankovic
- Department of Otolaryngology- Head and Neck Surgery, Stanford University School of Medicine, 801 Welch Rd, Palo Alto, Stanford, CA, 94304, USA.
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
5
|
Lander A, Kong Y, Jin Y, Wu C, Luk LYP. Deciphering the Synthetic and Refolding Strategy of a Cysteine-Rich Domain in the Tumor Necrosis Factor Receptor (TNF-R) for Racemic Crystallography Analysis and d-Peptide Ligand Discovery. ACS BIO & MED CHEM AU 2024; 4:68-76. [PMID: 38404743 PMCID: PMC10885103 DOI: 10.1021/acsbiomedchemau.3c00060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 02/27/2024]
Abstract
Many cell-surface receptors are promising targets for chemical synthesis because of their critical roles in disease development. This synthetic approach enables investigations by racemic protein crystallography and ligand discovery by mirror-image methodologies. However, due to their complex nature, the chemical synthesis of a receptor can be a significant challenge. Here, we describe the chemical synthesis and folding of a central, cysteine-rich domain of the cell-surface receptor tumor necrosis factor 1 which is integral to binding of the cytokine TNF-α, namely, TNFR-1 CRD2. Racemic protein crystallography at 1.4 Å confirmed that the native binding conformation was preserved, and TNFR-1 CRD2 maintained its capacity to bind to TNF-α (KD ≈ 7 nM). Encouraged by this discovery, we carried out mirror-image phage display using the enantiomeric receptor mimic and identified a d-peptide ligand for TNFR-1 CRD2 (KD = 1 μM). This work demonstrated that cysteine-rich domains, including the central domains, can be chemically synthesized and used as mimics for investigations.
Collapse
Affiliation(s)
- Alexander
J. Lander
- School
of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Yifu Kong
- Department
of Chemistry, College of Chemistry and Chemical Engineering, The MOE
Key Laboratory of Spectrochemical Analysis and Instrumentation, State
Key Laboratory of Physical Chemistry of Solid Surfaces, Xiamen University, Fujian Province 361005, China
| | - Yi Jin
- Manchester
Institute of Biotechnology, University of
Manchester, Manchester M1 7DN, U.K.
| | - Chuanliu Wu
- Department
of Chemistry, College of Chemistry and Chemical Engineering, The MOE
Key Laboratory of Spectrochemical Analysis and Instrumentation, State
Key Laboratory of Physical Chemistry of Solid Surfaces, Xiamen University, Fujian Province 361005, China
| | - Louis Y. P. Luk
- School
of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| |
Collapse
|
6
|
Bessaad M, Habel A, Hadj Ahmed M, Xu W, Stayoussef M, Bouaziz H, Hachiche M, Mezlini A, Larbi A, Yaacoubi-Loueslati B. Assessing serum cytokine profiles in inflammatory breast cancer patients using Luminex® technology. Cytokine 2023; 172:156409. [PMID: 37918053 DOI: 10.1016/j.cyto.2023.156409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/03/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Inflammatory breast cancer (IBC), accounts for the majority of deaths associated with breast tumors. Because this form is aggressive from its appearance and has a strong metastatic potential. The majority of patients are not diagnosed until late stages, highlighting the need for the development of novel diagnostic biomarkers. Immune mediators may affect IBC progression and metastasis installation. AIM OF THE STUDY Analysis of serum proteins to identify a panel of prognostic biomarkers for IBC. PATIENTS AND METHODS Serum levels of 65 analytes were determined in IBC and Non-IBC patients with the ProcartaPlex Human Immune Monitoring 65-Plex Panel. RESULTS Fifteen analytes: 5 cytokines (IL-8, IL-16, IL-21, IL-22 and MIF), 7 chemokines (Eotaxin, eotaxin-3, Fractalkine, IP-10, MIP-1α, MIP-1β and SDF-1α), One growth factors (FGF-2) and 2 soluble receptors (TNFRII and Tweak); were significantly differentially expressed between the two groups. ROC curves showed that twelve of them (IL-8, IL-16, IL-21, IL-22, MIF, MIP-1α, MIP-1β, SDF-1α, TNFRII, FGF-2, Eotaxin-3, and Fractalkine) had AUC values greater than 0.70 and thus had potential clinical utility. Moreover, seven cytokines: IL-8, IL-16, MIF, Eotaxin-3, MIP-1α, MIP-1β, and CD-30 are positively associated with patients who developed distant metastasis. Ten analytes: Eotaxin-3, Fractalkine, IL-16, IL-1α, IL-22, IL-8, MIF, MIP-1α, MIP-1β, and TNFRII are positively associated with patients who had Lymph-Nodes invasion. CONCLUSION This study has uncovered a set of 8 analytes (Eotaxin-3, Fractalkine, IL-16, IL-8, IL-22, MIF, MIP-1α, MIP-1β) that can be used as biomarkers of IBC, and can be utilized for early detection of IBC, preventing metastasis and lymph-Nodes invasion.
Collapse
Affiliation(s)
- Maryem Bessaad
- University of Tunis El Manar (UTM), Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Tunisia
| | - Azza Habel
- University of Tunis El Manar (UTM), Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Tunisia
| | - Mariem Hadj Ahmed
- University of Tunis El Manar (UTM), Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Tunisia
| | - Weili Xu
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, Singapore 138648, Singapore
| | - Mouna Stayoussef
- University of Tunis El Manar (UTM), Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Tunisia
| | - Hanen Bouaziz
- Salah Azaiez Oncology Institute, Avenue 9 April, 1006, Bab Saadoun, Tunis, Tunisia
| | - Monia Hachiche
- Salah Azaiez Oncology Institute, Avenue 9 April, 1006, Bab Saadoun, Tunis, Tunisia
| | - Amel Mezlini
- Salah Azaiez Oncology Institute, Avenue 9 April, 1006, Bab Saadoun, Tunis, Tunisia
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, Singapore 138648, Singapore
| | - Besma Yaacoubi-Loueslati
- University of Tunis El Manar (UTM), Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), Tunisia.
| |
Collapse
|
7
|
Chen HC, Wang CW, Toh WH, Lee HE, Chung WH, Chen CB. Advancing Treatment in Bullous Pemphigoid: A Comprehensive Review of Novel Therapeutic Targets and Approaches. Clin Rev Allergy Immunol 2023; 65:331-353. [PMID: 37897588 DOI: 10.1007/s12016-023-08973-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2023] [Indexed: 10/30/2023]
Abstract
Bullous pemphigoid is one of the most common autoimmune bullous diseases occurring primarily in the elderly. Pathogenic autoantibodies against BP180 and BP230 at the dermal-epidermal junction cause subepidermal blisters, erosions, and intense pruritus, all of which adversely affect the patients' quality of life and may increase their morbidity and mortality. Current systemic treatment options for bullous pemphigoid are limited to corticosteroids and immunosuppressants, which can have substantial side effects on these vulnerable patients that even exceed their therapeutic benefits. Therefore, more precisely, targeting therapies to the pathogenic cells and molecules in bullous pemphigoid is an urgent issue. In this review, we describe the pathophysiology of bullous pemphigoid, focusing on autoantibodies, complements, eosinophils, neutrophils, proteases, and the T helper 2 and 17 axes since they are crucial in promoting proinflammatory environments. We also highlight the emerging therapeutic targets for bullous pemphigoid and their latest discoveries in clinical trials or experimental studies. Further well-designed studies are required to establish the efficacy and safety of these prospective therapeutic options.
Collapse
Affiliation(s)
- Hsuan-Chi Chen
- Department of Medical Education, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chuang-Wei Wang
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Linkou, Taipei, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Department of Medical Research, Chang Gung Memorial Hospital, Linkou, Taiwan
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China
- Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen, China
| | - Wu Han Toh
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Hua-En Lee
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Linkou, Taipei, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Department of Medical Research, Chang Gung Memorial Hospital, Linkou, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Hung Chung
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Linkou, Taipei, Taiwan.
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Department of Medical Research, Chang Gung Memorial Hospital, Linkou, Taiwan.
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung, Taiwan.
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China.
- Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen, China.
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan.
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan.
- Department of Dermatology, Beijing Tsinghua Chang Gung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China.
- Department of Dermatology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan.
| | - Chun-Bing Chen
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Linkou, Taipei, Taiwan.
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Department of Medical Research, Chang Gung Memorial Hospital, Linkou, Taiwan.
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung, Taiwan.
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China.
- Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen, China.
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan.
| |
Collapse
|
8
|
Siegmund D, Zaitseva O, Wajant H. Fn14 and TNFR2 as regulators of cytotoxic TNFR1 signaling. Front Cell Dev Biol 2023; 11:1267837. [PMID: 38020877 PMCID: PMC10657838 DOI: 10.3389/fcell.2023.1267837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Tumor necrosis factor (TNF) receptor 1 (TNFR1), TNFR2 and fibroblast growth factor-inducible 14 (Fn14) belong to the TNF receptor superfamily (TNFRSF). From a structural point of view, TNFR1 is a prototypic death domain (DD)-containing receptor. In contrast to other prominent death receptors, such as CD95/Fas and the two TRAIL death receptors DR4 and DR5, however, liganded TNFR1 does not instruct the formation of a plasma membrane-associated death inducing signaling complex converting procaspase-8 into highly active mature heterotetrameric caspase-8 molecules. Instead, liganded TNFR1 recruits the DD-containing cytoplasmic signaling proteins TRADD and RIPK1 and empowers these proteins to trigger cell death signaling by cytosolic complexes after their release from the TNFR1 signaling complex. The activity and quality (apoptosis versus necroptosis) of TNF-induced cell death signaling is controlled by caspase-8, the caspase-8 regulatory FLIP proteins, TRAF2, RIPK1 and the RIPK1-ubiquitinating E3 ligases cIAP1 and cIAP2. TNFR2 and Fn14 efficiently recruit TRAF2 along with the TRAF2 binding partners cIAP1 and cIAP2 and can thereby limit the availability of these molecules for other TRAF2/cIAP1/2-utilizing proteins including TNFR1. Accordingly, at the cellular level engagement of TNFR2 or Fn14 inhibits TNFR1-induced RIPK1-mediated effects reaching from activation of the classical NFκB pathway to induction of apoptosis and necroptosis. In this review, we summarize the effects of TNFR2- and Fn14-mediated depletion of TRAF2 and the cIAP1/2 on TNFR1 signaling at the molecular level and discuss the consequences this has in vivo.
Collapse
Affiliation(s)
| | | | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
9
|
Zaitseva O, Hoffmann A, Löst M, Anany MA, Zhang T, Kucka K, Wiegering A, Otto C, Wajant H. Antibody-based soluble and membrane-bound TWEAK mimicking agonists with FcγR-independent activity. Front Immunol 2023; 14:1194610. [PMID: 37545514 PMCID: PMC10402896 DOI: 10.3389/fimmu.2023.1194610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/22/2023] [Indexed: 08/08/2023] Open
Abstract
Fibroblast growth factor (FGF)-inducible 14 (Fn14) activates the classical and alternative NFκB (nuclear factor 'kappa-light-chain-enhancer' of activated B-cells) signaling pathway but also enhances tumor necrosis factor (TNF)-induced cell death. Fn14 expression is upregulated in non-hematopoietic cells during tissue injury and is also often highly expressed in solid cancers. In view of the latter, there were and are considerable preclinical efforts to target Fn14 for tumor therapy, either by exploiting Fn14 as a target for antibodies with cytotoxic activity (e.g. antibody-dependent cellular cytotoxicity (ADCC)-inducing IgG variants, antibody drug conjugates) or by blocking antibodies with the aim to interfere with protumoral Fn14 activities. Noteworthy, there are yet no attempts to target Fn14 with agonistic Fc effector function silenced antibodies to unleash the proinflammatory and cell death-enhancing activities of this receptor for tumor therapy. This is certainly not at least due to the fact that anti-Fn14 antibodies only act as effective agonists when they are presented bound to Fcγ receptors (FcγR). Thus, there are so far no antibodies that robustly and selectively engage Fn14 signaling without triggering unwanted FcγR-mediated activities. In this study, we investigated a panel of variants of the anti-Fn14 antibody 18D1 of different valencies and domain architectures with respect to their inherent FcγR-independent ability to trigger Fn14-associated signaling pathways. In contrast to conventional 18D1, the majority of 18D1 antibody variants with four or more Fn14 binding sites displayed a strong ability to trigger the alternative NFκB pathway and to enhance TNF-induced cell death and therefore resemble in their activity soluble (TNF)-like weak inducer of apoptosis (TWEAK), one form of the natural occurring ligand of Fn14. Noteworthy, activation of the classical NFκB pathway, which naturally is predominately triggered by membrane-bound TWEAK but not soluble TWEAK, was preferentially observed with a subset of constructs containing Fn14 binding sites at opposing sites of the IgG scaffold, e.g. IgG1-scFv fusion proteins. A superior ability of IgG1-scFv fusion proteins to trigger classical NFκB signaling was also observed with the anti-Fn14 antibody PDL192 suggesting that we identified generic structures for Fn14 antibody variants mimicking soluble and membrane-bound TWEAK.
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, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Margaretha Löst
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Mohamed A. Anany
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- Department of Microbial Biotechnology, Institute of Biotechnology, National Research Center, Giza, Egypt
| | - Tengyu Zhang
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Kirstin Kucka
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Armin Wiegering
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Christoph Otto
- Department of General, Visceral, Transplant, 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
| |
Collapse
|
10
|
Chronic Trypanosoma cruzi infection activates the TWEAK/Fn14 axis in cardiac myocytes and fibroblasts driving structural and functional changes that affect the heart. Exp Parasitol 2023; 248:108491. [PMID: 36841467 DOI: 10.1016/j.exppara.2023.108491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/04/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023]
Abstract
Sustained interaction between the cytokine tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its functional receptor, fibroblast growth factor-inducible 14 (Fn14), has been linked to cardiovascular disorders. Chagas cardiomyopathy, elicited by Trypanosoma cruzi infection, is associated with chronic inflammation, fibrosis and hypertrophy. This study aimed to explore the involvement of the TWEAK/Fn 14 axis in development of Chagas heart disease. Parasite infection in vitro triggered Fn14 overexpression in atrial HL-1 myocytes and cardiac MCF fibroblasts. Fn14 levels were also increased in heart tissue from C57BL/6 mice at 130 days post-infection, particularly in myocytes and fibroblasts. Concurrently, TWEAK expression in circulating monocytes from this group was higher than that determined in uninfected controls. TWEAK/Fn14 interaction was functional in myocytes and fibroblasts isolated from infected hearts, leading to TNF receptor-associated factor 2 (TRAF2)-mediated activation of nuclear factor kappa B (NFκB) signaling. Ex vivo stimulation of both cell types with recombinant TWEAK for 24 h boosted the NFκB-regulated production of proinflammatory/profibrotic mediators (IL-1β, IL-6, TNF-α, IL-8, CCL2, CCL5, MMP-2, MMP-9, ICAM-1, E-selectin) involved in chronic T. cruzi cardiomyopathy. We further evaluated the therapeutic potential of the soluble decoy receptor Fn14-Fc to interfere with TWEAK/Fn14-dependent pathogenic activity. Fn14-Fc treatment of chronically infected mice was effective in neutralizing the ligand and reverting electrocardiographic abnormalities, maladaptive inflammation, adverse remodeling and hypertrophy in myocardium. Altogether, these findings suggest that sustained TWEAK/Fn14 induction by persistent T. cruzi infection is implicated in cardiopathogenesis and make TWEAK/Fn14 axis a promising target for the treatment of chronic Chagas heart disease.
Collapse
|
11
|
Extracellular Vesicle-Associated TWEAK Contributes to Vascular Inflammation and Remodeling During Acute Cellular Rejection. JACC Basic Transl Sci 2023. [DOI: 10.1016/j.jacbts.2022.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
12
|
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
|
13
|
Diagnostic test accuracy of novel biomarkers for lupus nephritis-An overview of systematic reviews. PLoS One 2022; 17:e0275016. [PMID: 36215243 PMCID: PMC9550089 DOI: 10.1371/journal.pone.0275016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with multiorgan inflammatory involvement and a mortality rate that is 2.6-fold higher than individuals of the same age and sex in the general population. Approximately 50% of patients with SLE develop renal impairment (lupus nephritis). Delayed diagnosis of lupus nephritis is associated with a higher risk of progression to end-stage renal disease, the need for replacement therapy, and mortality. The initial clinical manifestations of lupus nephritis are often discrete or absent and are usually detected through complementary tests. Although widely used in clinical practice, their accuracy is limited. A great scientific effort has been exerted towards searching for new, more sensitive, and specific biomarkers in recent years. Some systematic reviews have individually evaluated new serum and urinary biomarkers tested in patients with lupus nephritis. This overview aimed to summarize systematic reviews on the accuracy of novel serum and urinary biomarkers for diagnosing lupus nephritis in patients with SLE, discussing how our results can guide the clinical management of the disease and the direction of research in this area. METHODS The research question is "What is the accuracy of the new serum and urinary biomarkers studied for the diagnosis of LN in patients with SLE?". We searched for systematic reviews of observational studies evaluating the diagnostic accuracy of new serum or urinary biomarkers of lupus nephritis. The following databases were included: PubMed, EMBASE, BIREME/LILACS, Scopus, Web of Science, and Cochrane, including gray literature found via Google Scholar and PROQUEST. Two authors assessed the reviews for inclusion, data extraction, and assessment of the risk of bias (ROBIS tool). RESULTS Ten SRs on the diagnostic accuracy of new serum and urinary BMs in LN were selected. The SRs evaluated 7 distinct BMs: (a) antibodies (anti-Sm, anti-RNP, and anti-C1q), (b) cytokines (TWEAK and MCP-1), (c) a chemokine (IP-10), and (d) an acute phase glycoprotein (NGAL), in a total of 20 review arms (9 that analyzed serum BMs, and 12 that analyzed BMs in urine). The population evaluated in the primary studies was predominantly adults. Two SRs included strictly adults, 5 reviews also included studies in the paediatric population, and 4 did not report the age groups. The results of the evaluation with the ROBIS tool showed that most of the reviews had a low overall risk of bias. CONCLUSIONS There are 10 SRs of evidence relating to the diagnostic accuracy of serum and urinary biomarkers for lupus nephritis. Among the BMs evaluated, anti-C1q, urinary MCP-1, TWEAK, and NGAL stood out, highlighting the need for additional research, especially on LN diagnostic panels, and attempting to address methodological issues within diagnostic accuracy research. This would allow for a better understanding of their usefulness and possibly validate their clinical use in the future. REGISTRATION This project is registered on the International Prospective Registry of Systematic Reviews (PROSPERO) database (CRD42020196693).
Collapse
|
14
|
Güner G, Aßfalg M, Zhao K, Dreyer T, Lahiri S, Lo Y, Slivinschi BI, Imhof A, Jocher G, Strohm L, Behrends C, Langosch D, Bronger H, Nimsky C, Bartsch JW, Riddell SR, Steiner H, Lichtenthaler SF. Proteolytically generated soluble Tweak Receptor Fn14 is a blood biomarker for γ-secretase activity. EMBO Mol Med 2022; 14:e16084. [PMID: 36069059 PMCID: PMC9549706 DOI: 10.15252/emmm.202216084] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/29/2022] [Accepted: 08/15/2022] [Indexed: 11/12/2022] Open
Abstract
Fn14 is a cell surface receptor with key functions in tissue homeostasis and injury but is also linked to chronic diseases. Despite its physiological and medical importance, the regulation of Fn14 signaling and turnover is only partly understood. Here, we demonstrate that Fn14 is cleaved within its transmembrane domain by the protease γ‐secretase, resulting in secretion of the soluble Fn14 ectodomain (sFn14). Inhibition of γ‐secretase in tumor cells reduced sFn14 secretion, increased full‐length Fn14 at the cell surface, and enhanced TWEAK ligand‐stimulated Fn14 signaling through the NFκB pathway, which led to enhanced release of the cytokine tumor necrosis factor. γ‐Secretase‐dependent sFn14 release was also detected ex vivo in primary tumor cells from glioblastoma patients, in mouse and human plasma and was strongly reduced in blood from human cancer patients dosed with a γ‐secretase inhibitor prior to chimeric antigen receptor (CAR)‐T‐cell treatment. Taken together, our study demonstrates a novel function for γ‐secretase in attenuating TWEAK/Fn14 signaling and suggests the use of sFn14 as an easily measurable pharmacodynamic biomarker to monitor γ‐secretase activity in vivo.
Collapse
Affiliation(s)
- Gökhan Güner
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Marlene Aßfalg
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Kai Zhao
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Tobias Dreyer
- Department of Gynecology and Obstetrics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Shibojyoti Lahiri
- Protein Analysis Unit, Faculty of Medicine, Biomedical Center, LMU, Martinsried, Germany
| | - Yun Lo
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Bianca Ionela Slivinschi
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Axel Imhof
- Protein Analysis Unit, Faculty of Medicine, Biomedical Center, LMU, Martinsried, Germany
| | - Georg Jocher
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Laura Strohm
- Munich Cluster for Systems Neurology (SyNergy), Medical Faculty, LMU, Munich, Germany
| | - Christian Behrends
- Munich Cluster for Systems Neurology (SyNergy), Medical Faculty, LMU, Munich, Germany
| | | | - Holger Bronger
- Department of Gynecology and Obstetrics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christopher Nimsky
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Jörg W Bartsch
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Stanley R Riddell
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Harald Steiner
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Division of Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), LMU, Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
15
|
A novel shiga based immunotoxin against Fn-14 receptor on colorectal and lung cancer. Int Immunopharmacol 2022; 110:109076. [DOI: 10.1016/j.intimp.2022.109076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/28/2022] [Accepted: 07/18/2022] [Indexed: 11/20/2022]
|
16
|
3β-Hydroxy-5β-hydroxy-B-norcholestane-6β-carboxaldehyde (SEC-B) Induces Proinflammatory Activation of Human Endothelial Cells Associated with Nitric Oxide Production and Endothelial Nitric Oxide Synthase/Caveolin-1 Dysregulation. Antioxidants (Basel) 2022; 11:antiox11061148. [PMID: 35740044 PMCID: PMC9220016 DOI: 10.3390/antiox11061148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022] Open
Abstract
Oxysterols are a family of 27-carbon cholesterol oxidation derivatives found in low-density lipoproteins (LDLs) and atherosclerotic plaques where they trigger several biological responses involved in the initiation and progression of atherosclerosis. Several pieces of evidence suggest that oxysterols contribute to endothelial dysfunction (ED) due to their ability to alter membrane fluidity and cell permeability leading to inflammation, oxidative stress and apoptosis. The present study aimed to investigate the molecular events occurring in human microvascular endothelial cells (HMEC-1) in response to autoxidation-generated 3β-hydroxy-5β-hydroxy-B-norcholestane-6β-carboxaldehyde (SEC-B) exposure. Our results highlight that SEC-B rapidly activates HMEC-1 by inducing oxidative stress, nitric oxide (NO) production and pro-inflammatory cytokine release. Exposure to SEC-B up to 24 h results in persistent accumulation of the vasodilator NO paralleled by an upregulation of the endothelial nitric oxide synthase (eNOS) enzyme and downregulation of Caveolin-1 (Cav-1) protein levels. Moreover, reduced expression and extracellular release of the vasoconstrictor factor endothelin-1 (ET-1) are observed. Furthermore, SEC-B stimulates the expression of the cytokines interleukin-6 (IL-6) and tumor necrosis factor-like weak inducer of apoptosis (TWEAK). This proinflammatory state leads to increased monocyte recruitment on activated HMEC-1 cells. Our findings add new knowledge on the role of SEC-B in ED and further support its potential implication in atherosclerosis.
Collapse
|
17
|
Chan JCY, Gorski SM. Unlocking the gate to GABARAPL2. Biol Futur 2022; 73:157-169. [PMID: 35486231 DOI: 10.1007/s42977-022-00119-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
Abstract
GABARAPL2 was initially characterized for its involvement in protein transport and membrane fusion events, but has since gained notoriety for its role in autophagy. GABARAPL2 is frequently studied alongside its GABARAP subfamily members, GABARAP and GABARAPL1. Although functional redundancy exists among the subfamily members, a complex network of molecular interactions, physiological processes and pathologies can be primarily related to GABARAPL2. GABARAPL2 has a multifaceted role, ranging from cellular differentiation to intracellular degradation. Much of what we know about GABARAPL2 is gained through identifying its interacting partners-a list that is constantly growing. In this article, we review both the autophagy-dependent and autophagy-independent roles of GABARAPL2, and emphasize their implications for both health and disease.
Collapse
Affiliation(s)
- Jennifer C Y Chan
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada.,Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, V5Z 1L3, Canada
| | - Sharon M Gorski
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada. .,Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, V5Z 1L3, Canada. .,Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada.
| |
Collapse
|
18
|
Silva‐Candal A, Custodia A, López‐Dequidt I, Rodríguez‐Yáñez M, Alonso‐Alonso ML, Ávila‐Gómez P, Pumar JM, Castillo J, Sobrino T, Campos F, Iglesias‐Rey R, Hervella P. sTWEAK
is a leukoaraiosis biomarker associated with neurovascular angiopathy. Ann Clin Transl Neurol 2022; 9:171-180. [PMID: 35060359 PMCID: PMC8862435 DOI: 10.1002/acn3.51502] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/15/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Objective Leukoaraiosis (LA) refers to white matter lesions of undetermined etiology associated with the appearance and worsening of vascular pathologies. The aim is to confirm an increased frequency and intensity of LA in symptomatic patients with neurovascular pathology compared with asymptomatic subjects, and its association with circulating serum levels of soluble tumor necrosis factor‐like weak inducer of apoptosis (sTWEAK). Methods An observational study was conducted in which two groups of patients were compared. Group I (N = 242) comprised of asymptomatic subjects with arterial hypertension and/or diabetes or with a history of transient ischemic attacks, and Group II (N = 382) comprised patients with lacunar stroke or deep hemispheric intracerebral hemorrhage (ICH) of hypertensive origin. Serum levels of sTWEAK were analyzed and correlated with prevalence and intensity of LA according to the Fazekas scale. Results The prevalence of LA was higher in symptomatic (85.1%) versus asymptomatic patients (62.0%). Logistic regression model showed a significant relation of LA with neurovascular pathologies (OR: 2.69, IC 95%: 1.10–6.59, p = 0.003). When stratified according to the Fazekas scale, LA of grade II (OR: 3.53, IC 95%: 1.10–6.59, p = 0.003) and specially grade III (OR: 4.66, 95% CI: 1.09–19.84, p = 0.037) showed correlation with neurovascular pathologies. Increased sTWEAK levels were found in the symptomatic group in all LA grades (p < 0.0001), and associated with 5.06 times more risk of presenting clinical symptoms (OR: 5.06, 95% CI: 2.66–9.75, p < 0.0001). Interpretation LA showed a higher prevalence in patients with symptomatic lacunar stroke or deep hemispheric ICH. There is an association between sTWEAK levels and LA degree.
Collapse
Affiliation(s)
- Andrés Silva‐Candal
- Clinical Neurosciences Research Laboratories (LINC) Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
- Neurovascular Diseases Laboratory Neurology Service University Hospital Complex of A Coruña Biomedical Research Institute (INIBIC) A Coruña Spain
| | - Antia Custodia
- Clinical Neurosciences Research Laboratories (LINC) Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - Iria López‐Dequidt
- Stroke Unit Department of Neurology Hospital Clínico Universitario Santiago de Compostela Spain
| | - Manuel Rodríguez‐Yáñez
- Stroke Unit Department of Neurology Hospital Clínico Universitario Santiago de Compostela Spain
| | - Maria Luz Alonso‐Alonso
- Clinical Neurosciences Research Laboratories (LINC) Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - Paulo Ávila‐Gómez
- Clinical Neurosciences Research Laboratories (LINC) Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - José M. Pumar
- Department of Neuroradiology Hospital Clínico Universitario Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratories (LINC) Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratories (LINC) Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratories (LINC) Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - Ramón Iglesias‐Rey
- Clinical Neurosciences Research Laboratories (LINC) Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| | - Pablo Hervella
- Clinical Neurosciences Research Laboratories (LINC) Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
| |
Collapse
|
19
|
Ding S, Li H, Zhang YH, Zhou X, Feng K, Li Z, Chen L, Huang T, Cai YD. Identification of Pan-Cancer Biomarkers Based on the Gene Expression Profiles of Cancer Cell Lines. Front Cell Dev Biol 2021; 9:781285. [PMID: 34917619 PMCID: PMC8669964 DOI: 10.3389/fcell.2021.781285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022] Open
Abstract
There are many types of cancers. Although they share some hallmarks, such as proliferation and metastasis, they are still very different from many perspectives. They grow on different organ or tissues. Does each cancer have a unique gene expression pattern that makes it different from other cancer types? After the Cancer Genome Atlas (TCGA) project, there are more and more pan-cancer studies. Researchers want to get robust gene expression signature from pan-cancer patients. But there is large variance in cancer patients due to heterogeneity. To get robust results, the sample size will be too large to recruit. In this study, we tried another approach to get robust pan-cancer biomarkers by using the cell line data to reduce the variance. We applied several advanced computational methods to analyze the Cancer Cell Line Encyclopedia (CCLE) gene expression profiles which included 988 cell lines from 20 cancer types. Two feature selection methods, including Boruta, and max-relevance and min-redundancy methods, were applied to the cell line gene expression data one by one, generating a feature list. Such list was fed into incremental feature selection method, incorporating one classification algorithm, to extract biomarkers, construct optimal classifiers and decision rules. The optimal classifiers provided good performance, which can be useful tools to identify cell lines from different cancer types, whereas the biomarkers (e.g. NCKAP1, TNFRSF12A, LAMB2, FKBP9, PFN2, TOM1L1) and rules identified in this work may provide a meaningful and precise reference for differentiating multiple types of cancer and contribute to the personalized treatment of tumors.
Collapse
Affiliation(s)
- ShiJian Ding
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Hao Li
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Yu-Hang Zhang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - XianChao Zhou
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - KaiYan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou, China
| | - ZhanDong Li
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Tao Huang
- CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
20
|
Abós B, Pérez-Fernández E, Morel E, Perdiguero P, Tafalla C. Pro-Inflammatory and B Cell Regulating Capacities of TWEAK in Rainbow Trout ( Oncorhynchus mykiss). Front Immunol 2021; 12:748836. [PMID: 34659247 PMCID: PMC8517431 DOI: 10.3389/fimmu.2021.748836] [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: 07/28/2021] [Accepted: 09/15/2021] [Indexed: 11/22/2022] Open
Abstract
Tumor necrosis factor (TNF)-like weak inducer of apoptosis or TWEAK is a member of the TNF superfamily involved in the regulation of many biological processes. In mammals, TWEAK has been shown to play a role in some autoimmune or inflammatory conditions, but its immune role is not yet clearly defined. In teleost fish, although a few studies have identified homologues to mammalian TWEAK, their biological effects have never been investigated. In the current study, we have studied the transcriptional regulation of two TWEAK homologues (TWEAK 1 and 2) identified in rainbow trout (Oncorhynchus mykiss) throughout different tissues, in response to parasitic or viral infections, or in head kidney (HK) leukocytes stimulated with different stimuli. Although the transcription of both homologues was modulated when HK leukocytes were exposed to several immune stimuli, only TWEAK 1 was significantly modulated upon pathogenic exposure. Thus, we performed a characterization of the functions exerted by this cytokine in HK leukocytes. Recombinant TWEAK 1 strongly up-regulated the transcription of pro-inflammatory genes and antimicrobial peptides in HK leukocytes, with differential transcriptional effects in IgM+ B cells, IgM- lymphocytes and myeloid cells. TWEAK 1 also increased the survival and promoted the differentiation of B cells in HK leukocyte cultures. Our results demonstrate that in teleost fish, TWEAK 1 is involved in the response to different types of pathogens, through the modulation of antimicrobial and pro-inflammatory genes in different leukocytes subsets. Furthermore, a role for TWEAK as a B cell differentiation factor has also been established in rainbow trout.
Collapse
Affiliation(s)
- Beatriz Abós
- Animal Health Research Center (CISA), Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Elena Pérez-Fernández
- Animal Health Research Center (CISA), Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Esther Morel
- Animal Health Research Center (CISA), Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Pedro Perdiguero
- Animal Health Research Center (CISA), Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carolina Tafalla
- Animal Health Research Center (CISA), Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
21
|
Aido A, Zaitseva O, Wajant H, Buzgo M, Simaite A. Anti-Fn14 Antibody-Conjugated Nanoparticles Display Membrane TWEAK-Like Agonism. Pharmaceutics 2021; 13:pharmaceutics13071072. [PMID: 34371763 PMCID: PMC8308961 DOI: 10.3390/pharmaceutics13071072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 11/26/2022] Open
Abstract
Conventional bivalent IgG antibodies targeting a subgroup of receptors of the TNF superfamily (TNFSF) including fibroblast growth factor-inducible 14 (anti-Fn14) typically display no or only very limited agonistic activity on their own and can only trigger receptor signaling by crosslinking or when bound to Fcγ receptors (FcγR). Both result in proximity of multiple antibody-bound TNFRSF receptor (TNFR) molecules, which enables engagement of TNFR-associated signaling pathways. Here, we have linked anti-Fn14 antibodies to gold nanoparticles to mimic the “activating” effect of plasma membrane-presented FcγR-anchored anti-Fn14 antibodies. We functionalized gold nanoparticles with poly-ethylene glycol (PEG) linkers and then coupled antibodies to the PEG surface of the nanoparticles. We found that Fn14 binding of the anti-Fn14 antibodies PDL192 and 5B6 is preserved upon attachment to the nanoparticles. More importantly, the gold nanoparticle-presented anti-Fn14 antibody molecules displayed strong agonistic activity. Our results suggest that conjugation of monoclonal anti-TNFR antibodies to gold nanoparticles can be exploited to uncover their latent agonism, e.g., for immunotherapeutic applications.
Collapse
Affiliation(s)
- Ahmed Aido
- InoCure s.r.o, Department of R&D, Prumyslová 1960, 250 88 Celákovice, Czech Republic; (M.B.); (A.S.)
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Auverahaus, Grombühlstrasse 12, 97080 Würzburg, Germany; (O.Z.); (H.W.)
- Correspondence: ; Tel.: +49-174-913-6442
| | - Olena Zaitseva
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Auverahaus, Grombühlstrasse 12, 97080 Würzburg, Germany; (O.Z.); (H.W.)
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Auverahaus, Grombühlstrasse 12, 97080 Würzburg, Germany; (O.Z.); (H.W.)
| | - Matej Buzgo
- InoCure s.r.o, Department of R&D, Prumyslová 1960, 250 88 Celákovice, Czech Republic; (M.B.); (A.S.)
| | - Aiva Simaite
- InoCure s.r.o, Department of R&D, Prumyslová 1960, 250 88 Celákovice, Czech Republic; (M.B.); (A.S.)
| |
Collapse
|
22
|
Hervella P, Pérez-Mato M, Rodríguez-Yáñez M, López-Dequidt I, Pumar JM, Sobrino T, Campos F, Castillo J, da Silva-Candal A, Iglesias-Rey R. sTWEAK as Predictor of Stroke Recurrence in Ischemic Stroke Patients Treated With Reperfusion Therapies. Front Neurol 2021; 12:652867. [PMID: 34046003 PMCID: PMC8144448 DOI: 10.3389/fneur.2021.652867] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/09/2021] [Indexed: 11/20/2022] Open
Abstract
Aim: The purpose of this study was to investigate clinical and neuroimaging factors associated with stroke recurrence in reperfused ischemic stroke patients, as well as the influence of specific biomarkers of inflammation and endothelial dysfunction. Methods: We conducted a retrospective analysis on a prospectively registered database. Of the 875 patients eligible for this study (53.9% males; mean age 69.6 ± 11.8 years vs. 46.1% females; mean age 74.9 ± 12.6 years), 710 underwent systemic thrombolysis, 87 thrombectomy and in 78, systemic or intra-arterial thrombolysis together with thrombectomy was applied. Plasma levels of interleukin 6 (IL-6) and tumor necrosis factor alpha (TNFα) were analyzed as markers of inflammation, and soluble tumor necrosis factor-like inducer of apoptosis (sTWEAK) as an endothelial dysfunction marker. The main outcome variables of the study were the presence and severity of leukoaraiosis (LA) and stroke recurrence. Results: The average follow-up time of the study was 25 ± 13 months, during which 127 patients (14.5%) showed stroke recurrence. The presence and severity of LA was more severe in the second stroke episode (Grade III of the Fazekas 28.3 vs. 52.8%; p < 0.0001). IL-6 levels at the first admission and before reperfusion treatment in patients with and without subsequent recurrence were similar (9.9 ± 10.4 vs. 9.1 ± 7.0 pg/mL, p = 0.439), but different for TNFα (14.7 ± 5.6 vs. 15.9 ± 5.7 pg/mL, p = 0.031) and sTWEAK (5,970.8 ± 4,330.4 vs. 8,660.7 ± 5,119.0 pg/mL, p < 0.0001). sTWEAK values ≥7,000 pg/mL determined in the first stroke were independently associated to recurrence (OR 2.79; CI 95%: 1.87–4.16, p < 0.0001). Conclusions: The severity and the progression of LA are the main neuroimaging factors associated with stroke recurrence. Likewise, sTWEAK levels were independently associated to stroke recurrence, so further studies are necessary to investigate sTWEAK as a therapeutic target.
Collapse
Affiliation(s)
- Pablo Hervella
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - María Pérez-Mato
- Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital, IdiPAZ, UAM, Madrid, Spain
| | - Manuel Rodríguez-Yáñez
- Stroke Unit, Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela, Spain
| | - Iria López-Dequidt
- Stroke Unit, Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela, Spain
| | - José M Pumar
- Department of Neuroradiology, Hospital Clínico Universitario, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Andrés da Silva-Candal
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
23
|
da Silva-Candal A, López-Dequidt I, Rodriguez-Yañez M, Ávila-Gómez P, Pumar JM, Castillo J, Sobrino T, Campos F, Iglesias-Rey R, Hervella P. sTWEAK is a marker of early haematoma growth and leukoaraiosis in intracerebral haemorrhage. Stroke Vasc Neurol 2021; 6:528-535. [PMID: 33758070 PMCID: PMC8717766 DOI: 10.1136/svn-2020-000684] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/24/2020] [Accepted: 01/16/2021] [Indexed: 11/17/2022] Open
Abstract
Objective To study the association between early growth of haematoma with biomarkers of endothelial dysfunction such as leukoaraiosis (LA) and the soluble tumour necrosis factor-like weak inducer of apoptosis (sTWEAK) in patients with intracerebral haemorrhage (ICH). Methods This is a retrospective observational study of patients with nontraumatic ICH. Clinical and biochemical parameters were analysed. sTWEAK levels were measured by ELISA. LA was analysed in the hemisphere without haemorrhage to avoid interference with the acute injury. The main endpoint was the haematoma growth evaluated by the difference in volume between the second and the initial neuroimage. Poor functional outcome, defined as a modified Rankin Scale >2 at 3 months, was considered as secondary endpoint. Receiver operating characteristic curve analysis was performed to stablish the best cut-off for sTWEAK levels associated with haematoma growth. Results We included 653 patients with ICH in our analysis (71.1±11.9 years, 44% women). Haematoma growth was observed in 188 patients (28.8%). sTWEAK levels ≥5600 pg/mL predicted ICH growth with a sensitivity of 84% and a specificity of 87%. sTWEAK levels ≥5600 pg/mL and the presence of LA were associated with haematoma growth (OR: 42.46; (CI 95% 22.67 to 79.52) and OR: 2.73 (CI 95% 1.39 to 5.34), respectively). Also, the presence of LA (OR: 4.31 (CI 95% 2.89 to 6.42)) and the interaction between ICH growth and sTWEAK (OR: 2.23 (CI 95% 1.40 to 3.55)) were associated with poor functional outcome at 3 months. Conclusion sTWEAKs, together with the presence and grade of LA, are biomarkers able to predict ICH growth and poor functional outcome in patients with ICH.
Collapse
Affiliation(s)
- Andrés da Silva-Candal
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Iria López-Dequidt
- Department of Neurology, Hospital Clínico Universitario, Santiago de Compostela, Spain
| | | | - Paulo Ávila-Gómez
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - José Manuel Pumar
- Department of Neuroradiology, Hospital Clínico Universitario, Santiago de Compostela, Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Pablo Hervella
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
24
|
Eid A, Issa Y, Mohamed A, Badran F. Interleukin-9 and soluble tumor necrosis factor-like weak inducer of apoptosis in serum and suction blister fluid of nonsegmental vitiligo patients: Relation to disease severity. DERMATOL SIN 2021. [DOI: 10.4103/ds.ds_44_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
25
|
da Silva-Candal A, Pérez-Mato M, Rodríguez-Yáñez M, López-Dequidt I, Pumar JM, Ávila-Gómez P, Sobrino T, Campos F, Castillo J, Hervella P, Iglesias-Rey R. The presence of leukoaraiosis enhances the association between sTWEAK and hemorrhagic transformation. Ann Clin Transl Neurol 2020; 7:2103-2114. [PMID: 33022893 PMCID: PMC7664267 DOI: 10.1002/acn3.51171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/04/2020] [Accepted: 08/09/2020] [Indexed: 12/17/2022] Open
Abstract
Objective To investigate whether elevated serum levels of sTWEAK (soluble tumor necrosis factor‐like inducer of apoptosis) might be involved in a higher frequency of symptomatic hemorrhagic transformation (HT) through the presence of leukoaraiosis (LA) in patients with acute ischemic stroke (IS) undergoing reperfusion therapies. Methods This is a retrospective observational study. The primary endpoint was to study the sTWEAK‐LA‐HT relationship by comparing results with biomarkers associated to HT and evaluating functional outcome at 3‐months. Clinical factors, neuroimaging variables and biomarkers associated to inflammation, endothelial/atrial dysfunction or blood‐brain barrier damage were also investigated. Results We enrolled 875 patients (mean age 72.3 ± 12.2 years; 46.0% women); 710 individuals underwent intravenous thrombolysis, 87 endovascular therapy and 78 both. HT incidence was 32%; LA presence was 75.4%. Patients with poor functional outcome at 3‐months showed higher sTWEAK levels at admission (9844.2 [7460.4–12,542.0] vs. 2717.3 [1489.7–5852.3] pg/mL, P < 0.0001). By means of logistic regression models, PDGF‐CC and sTWEAK were associated with mechanisms linked simultaneously to HT and LA. Serum sTWEAK levels at admission ≥6700 pg/mL were associated with an odds ratio of 13 for poor outcome at 3‐months (OR: 13.6; CI 95%: 8.2–22.6, P < 0.0001). Conclusions Higher sTWEAK levels are independently associated with HT and poor functional outcome in patients with IS undergoing reperfusion therapies through the presence of LA. sTWEAK could become a therapeutic target to reduce HT incidence in patients with IS.
Collapse
Affiliation(s)
- Andrés da Silva-Candal
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - María Pérez-Mato
- Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital, IdiPAZ, UAM, Paseo de la Castellana 261, Madrid, 28046, Spain
| | - Manuel Rodríguez-Yáñez
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, Santiago de Compostela, Spain
| | - Iria López-Dequidt
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, Santiago de Compostela, Spain
| | - José M Pumar
- Department of Neuroradiology, Hospital Clínico Universitario, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Paulo Ávila-Gómez
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Pablo Hervella
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
26
|
Zhang Y, Zeng W, Xia Y. TWEAK/Fn14 axis is an important player in fibrosis. J Cell Physiol 2020; 236:3304-3316. [PMID: 33000480 DOI: 10.1002/jcp.30089] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022]
Abstract
Fibrosis is a common pathological condition associated with abnormal repair after tissue injury. However, the etiology and molecular mechanisms of fibrosis are still not well-understood. Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) belongs to the TNF superfamily and acts by binding to its receptor, fibroblast growth factor-inducible 14 (Fn14), thereby activating a variety of intracellular signal transduction pathways in various types of cells. Besides promoting the expression of growth factors, activation of TWEAK/Fn14 signaling after tissue injury can promote the expression of pro-inflammatory cytokines, which trigger the immune response, thereby exacerbating the injury. Severe or repetitive injury leads to a dysregulated tissue repair process, in which the TWEAK/Fn14 axis promotes the activation and proliferation of myofibroblasts, induces the secretion of the extracellular matrix, and regulates profibrotic mediators to further perpetuate and sustain the fibrotic process. In this review, we summarize the available experimental evidence on the underlying molecular mechanisms by which the TWEAK/Fn14 pathway mediates the development and progression of fibrosis. In addition, we discuss the therapeutic potential of the TWEAK/Fn14 pathway in fibrosis-associated diseases based on evidence derived from multiple models and cells from injured tissue and fibrotic tissue.
Collapse
Affiliation(s)
- Yitian Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Weihui Zeng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
27
|
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
|
28
|
Pascoe AL, Johnston AJ, Murphy RM. Controversies in TWEAK-Fn14 signaling in skeletal muscle atrophy and regeneration. Cell Mol Life Sci 2020; 77:3369-3381. [PMID: 32200423 PMCID: PMC11104974 DOI: 10.1007/s00018-020-03495-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/27/2020] [Accepted: 02/24/2020] [Indexed: 12/13/2022]
Abstract
Skeletal muscle is one of the largest functional tissues in the human body; it is highly plastic and responds dramatically to anabolic and catabolic stimuli, including weight training and malnutrition, respectively. Excessive loss of muscle mass, or atrophy, is a common symptom of many disease states with severe impacts on prognosis and quality of life. TNF-like weak inducer of apoptosis (TWEAK) and its cognate receptor, fibroblast growth factor-inducible 14 (Fn14) are an emerging cytokine signaling pathway in the pathogenesis of muscle atrophy. Upregulation of TWEAK and Fn14 has been described in a number of atrophic and injured muscle states; however, it remains unclear whether they are contributing to the degenerative or regenerative aspect of muscle insults. The current review focuses on the expression and apparent downstream outcomes of both TWEAK and Fn14 in a range of catabolic and anabolic muscle models. Apparent changes in the signaling outcomes of TWEAK-Fn14 activation dependent on the relative expression of both the ligand and the receptor are discussed as a potential source of divergent TWEAK-Fn14 downstream effects. This review proposes both a physiological and pathological model of TWEAK-Fn14 signaling. Further research is needed on the switch between these states to develop therapeutic interventions for this pathway.
Collapse
Affiliation(s)
- Amy L Pascoe
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Amelia J Johnston
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
29
|
El-Taweel AEAI, Abdelrahman AMN, Sabry S, Salem RM. Serum TWEAK: A cutoff between segmental and nonsegmental vitiligo. J Cosmet Dermatol 2020; 20:1017-1021. [PMID: 32808442 DOI: 10.1111/jocd.13644] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND TWEAK/Fn14 is expressed in many tissues including the skin, playing an important role in many inflammatory, autoimmune, and neoplastic cutaneous disorders. AIMS To assess the serum levels of tumor necrosis factor-like weak inducer of apoptosis (TWEAK) in vitiligo patients. METHODS This case-control study included 100 subjects (50 vitiligo patients and 50 control subjects) recruited from Dermatology Outpatient Clinic, Benha University. All patients were subjected to complete cutaneous examination, to evaluate the clinical type, distribution and severity of vitiligo using the Vitiligo Area Scoring Index (VASI). RESULTS TWEAK serum levels were significantly higher in patients than in the control subjects (644.76 ± 688.93 vs 282.75 ± 125.67, respectively). Serum levels were significantly elevated in segmental versus nonsegmental vitiligo. Receiver operating characteristic (ROC) analysis revealed that TWEAK shows 80% sensitivity and 56.67% specificity in diagnosing vitiligo and 100% sensitivity and 80.09% specificity in differentiating segmental from nonsegmental vitiligo. CONCLUSION TWEAK may play a role in vitiligo pathogenesis. It may be used in the differentiation between segmental and nonsegmental vitiligo and represent a promising therapeutic target in vitiligo.
Collapse
|
30
|
Unudurthi SD, Nassal DM, Patel NJ, Thomas E, Yu J, Pierson CG, Bansal SS, Mohler PJ, Hund TJ. Fibroblast growth factor-inducible 14 mediates macrophage infiltration in heart to promote pressure overload-induced cardiac dysfunction. Life Sci 2020; 247:117440. [PMID: 32070706 DOI: 10.1016/j.lfs.2020.117440] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/07/2020] [Accepted: 02/14/2020] [Indexed: 12/18/2022]
Abstract
AIMS Heart failure (HF) is characterized by compromised cardiac structure and function. Previous work has identified a link between upregulation of pro-inflammatory cytokines and HF. Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) is a pro-inflammatory cytokine, which binds to fibroblast growth factor inducible 14 (Fn14), a ubiquitously expressed cell-surface receptor. The objective of this study was to investigate the role of TWEAK/Fn14 pathway in promoting cardiac inflammation under non ischemic stress conditions. MAIN METHODS Wild type (WT) and Fn14 knock out (Fn14-/-) mice were subjected to pressure overload [transaortic constriction (TAC)] for 1 or 6 weeks. A subset of WT TAC animals were treated with the Fn14 antagonist L524-0366. Cardiac function was measured by echocardiography. Cardiac fibrosis and macrophage infiltration were quantified using immunohistochemistry and flow cytometry, respectively. Cardiac fibroblasts were isolated for quantifying TWEAK-induced chemokine release. KEY FINDINGS Fn14-/- mice displayed improved cardiac function, reduced fibrosis and lower macrophage infiltration in heart compared to WT following TAC. L524-0366 mitigated maladaptive remodeling with TAC. TWEAK induced secretion of the pro-inflammatory chemokine, monocyte chemoattractant protein 1 from WT but not Fn14-/- fibroblasts in vitro, in part through activation of non-canonical NF-κB signaling. Finally, Fn14 expression was increased in mouse following TAC and in human failing hearts. SIGNIFICANCE Our findings support an important role for the TWEAK/Fn14 promoting macrophage infiltration and fibrosis in heart under non-ischemic stress, with potential for therapeutic intervention to improve cardiac function in the setting of HF.
Collapse
Affiliation(s)
- Sathya D Unudurthi
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA.
| | - Drew M Nassal
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Nehal J Patel
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Evelyn Thomas
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Jane Yu
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Curtis G Pierson
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Shyam S Bansal
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology & Cell Biology, USA
| | - Peter J Mohler
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology & Cell Biology, USA; Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Thomas J Hund
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA; Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
31
|
Molecular Mode of Action of TRAIL Receptor Agonists-Common Principles and Their Translational Exploitation. Cancers (Basel) 2019; 11:cancers11070954. [PMID: 31284696 PMCID: PMC6678900 DOI: 10.3390/cancers11070954] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its death receptors TRAILR1/death receptor 4 (DR4) and TRAILR2/DR5 trigger cell death in many cancer cells but rarely exert cytotoxic activity on non-transformed cells. Against this background, a variety of recombinant TRAIL variants and anti-TRAIL death receptor antibodies have been developed and tested in preclinical and clinical studies. Despite promising results from mice tumor models, TRAIL death receptor targeting has failed so far in clinical studies to show satisfying anti-tumor efficacy. These disappointing results can largely be explained by two issues: First, tumor cells can acquire TRAIL resistance by several mechanisms defining a need for combination therapies with appropriate sensitizing drugs. Second, there is now growing preclinical evidence that soluble TRAIL variants but also bivalent anti-TRAIL death receptor antibodies typically require oligomerization or plasma membrane anchoring to achieve maximum activity. This review discusses the need for oligomerization and plasma membrane attachment for the activity of TRAIL death receptor agonists in view of what is known about the molecular mechanisms of how TRAIL death receptors trigger intracellular cell death signaling. In particular, it will be highlighted which consequences this has for the development of next generation TRAIL death receptor agonists and their potential clinical application.
Collapse
|
32
|
Di Martino L, Osme A, Kossak-Gupta S, Pizarro TT, Cominelli F. TWEAK/Fn14 Is Overexpressed in Crohn's Disease and Mediates Experimental Ileitis by Regulating Critical Innate and Adaptive Immune Pathways. Cell Mol Gastroenterol Hepatol 2019; 8:427-446. [PMID: 31181286 PMCID: PMC6718944 DOI: 10.1016/j.jcmgh.2019.05.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/24/2019] [Accepted: 05/28/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Crohn's disease (CD) is a debilitating inflammatory disorder that affects more than 1.6 million people in North America alone. Members of the tumor necrosis factor superfamily are key regulators of intestinal inflammation; specifically, tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor, fibroblast growth factor-inducible 14 (Fn14), are involved in normal and pathologic tissue remodeling. Our aim was to determine the role of TWEAK/Fn14 in CD and a murine model of CD-like ileitis (ie, SAMP1/YitFc [SAMP] strain). METHODS SAMP mice deficient in Fn14 (SAMP × Fn14-/-) were developed and a detailed time-course study was performed evaluating ileal tissues by histology and stereomicroscopy, as well as quantitative polymerase chain reaction and NanoString technology (Seattle, WA). Reciprocal bone marrow chimeras were generated to assess the relevance of Fn14 in hematopoietic vs nonhematopoietic compartments. Surgically resected intestinal tissues and mucosal biopsy specimens from patients with CD, ulcerative colitis, and healthy controls were analyzed for the expression of TWEAK/Fn14 by quantitative polymerase chain reaction, Western blot, immunohistochemistry, and immunofluorescence. RESULTS SAMP × Fn14-/- showed a marked decrease in ileitis severity at 20 weeks of age compared with SAMP WT controls. Bone marrow chimeras showed that Fn14 was required in both hematopoietic and nonhematopoietic compartments for ileitis to develop. Transcriptome data showed multiple cellular pathways regulated by Fn14 signaling. Finally, increased expression of TWEAK and Fn14 was observed in tissue lesions from CD patients compared with ulcerative colitis and healthy controls. CONCLUSIONS TWEAK/Fn14 are up-regulated in CD, and also mediate experimental CD-like ileitis, by regulation of multiple innate and adaptive cellular pathways. Therefore, TWEAK/Fn14 may represent a novel therapeutic target for the treatment of small intestinal inflammation in CD.
Collapse
Affiliation(s)
- Luca Di Martino
- Division of Gastroenterology and Liver Disease, Case Western University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio
| | - Abdullah Osme
- Division of Gastroenterology and Liver Disease, Case Western University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio
| | - Sarah Kossak-Gupta
- Division of Gastroenterology and Liver Disease, Case Western University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio
| | - Theresa T Pizarro
- Division of Gastroenterology and Liver Disease, Case Western University School of Medicine, Cleveland, Ohio; Department of Pathology, Case Western University School of Medicine, Cleveland, Ohio
| | - Fabio Cominelli
- Division of Gastroenterology and Liver Disease, Case Western University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western University School of Medicine, Cleveland, Ohio; Department of Pathology, Case Western University School of Medicine, Cleveland, Ohio.
| |
Collapse
|
33
|
Chen TH, Ma GC, Lin WH, Lee DJ, Wu SH, Liao BY, Chen M, Lin LK. Genome-Wide Microarray Analysis Suggests Transcriptomic Response May Not Play a Major Role in High- to Low-Altitude Acclimation in Harvest Mouse ( Micromys minutus). Animals (Basel) 2019; 9:ani9030092. [PMID: 30871279 PMCID: PMC6466072 DOI: 10.3390/ani9030092] [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: 02/25/2019] [Revised: 03/09/2019] [Accepted: 03/10/2019] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Micromys minutus is a small rodent species that has a wide range of vertical distribution in Taiwan. By comparing the gene expression profile of the skeletal muscle tissues taken from individuals native to the high-altitude environment and those transferred to the low-altitude captive site, the Tnfrsf12a gene was demonstrated to have a differential expression pattern. Although this finding may be correlated with the altitude acclimation, the observation of only one gene transcript with significant alteration leads us to suggest that genetic response may not play a major role in altitude acclimation in M. minutus. Future comparative functional genomics studies involving other organ systems (in addition to skeletal muscles) and alarger sample size are warranted for better insight into the altitude acclimation of this small rodent species. Abstract The harvest mouse (Micromys minutus) is a small rodent species with a wide range of vertical distribution in Taiwan, extending from the sea level to 3100 m altitude. This species has recently suffered from habitat loss in high-altitude areas due to orchard cultivation, which may have resulted in mouse migration from high to low altitude. To investigate whether there is any physiological mechanism involved in altitude acclimation, rat cDNA microarray was used to compare transcriptomic patterns of the skeletal muscle tissues taken from individuals native to the high-altitude environment and those transferred to the low-altitude captive site. Of the 23,188 genes being analyzed, 47 (33 up-regulated and 14 down-regulated) were found to have differential expression (fold change > 4 or < −4, ANOVA p < 0.05). However, after multiple testing correction with a false discovery rate (FDR), only the result for Tnfrsf12a was found to be statistically significant (fold change = 13, FDR p < 0.05). The result was confirmed by quantitative polymerase chain reaction (q-PCR). The expression of Tnfrsf12a possibly relates to the skeletal muscle biology and thus can be correlated with altitude acclimation. However, finding only one gene transcript with significant alteration suggests that transcriptomic response may not play a major role in high- to low-altitude acclimation in harvest mouse.
Collapse
Affiliation(s)
- Tze-Ho Chen
- Department of Life Science, Tunghai University, Taichung 40704, Taiwan.
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua 50006, Taiwan.
| | - Gwo-Chin Ma
- Department of Genomic Medicine and Center for Medical Genetics, Changhua Christian Hospital, Changhua 50046, Taiwan.
- Department of Genomic Science and Technology, Changhua Christian Hospital Healthcare System,Changhua Christian Hospital, Changhua 50046, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, Central Taiwan University of Science and Technology, Taichung 40601, Taiwan.
| | - Wen-Hsiang Lin
- Department of Genomic Medicine and Center for Medical Genetics, Changhua Christian Hospital, Changhua 50046, Taiwan.
- Department of Genomic Science and Technology, Changhua Christian Hospital Healthcare System,Changhua Christian Hospital, Changhua 50046, Taiwan.
| | - Dong-Jay Lee
- Department of Genomic Medicine and Center for Medical Genetics, Changhua Christian Hospital, Changhua 50046, Taiwan.
- Department of Genomic Science and Technology, Changhua Christian Hospital Healthcare System,Changhua Christian Hospital, Changhua 50046, Taiwan.
| | - Sheng-Hai Wu
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
| | - Ben-Yang Liao
- Division of Biostatistics & Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan.
| | - Ming Chen
- Department of Life Science, Tunghai University, Taichung 40704, Taiwan.
- Department of Genomic Medicine and Center for Medical Genetics, Changhua Christian Hospital, Changhua 50046, Taiwan.
- Department of Genomic Science and Technology, Changhua Christian Hospital Healthcare System,Changhua Christian Hospital, Changhua 50046, Taiwan.
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei 10041, Taiwan.
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 10041, Taiwan.
- Department of Molecular Biotechnology, Da-Yeh University, Changhua 51591, Taiwan.
| | - Liang-Kong Lin
- Department of Life Science, Tunghai University, Taichung 40704, Taiwan.
| |
Collapse
|
34
|
Delgado ME, Brunner T. The many faces of tumor necrosis factor signaling in the intestinal epithelium. Genes Immun 2019; 20:609-626. [DOI: 10.1038/s41435-019-0057-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 12/26/2018] [Indexed: 01/15/2023]
|
35
|
Lee SJ, Kim J, Ko J, Lee EJ, Koh HJ, Yoon JS. Tumor necrosis factor-like weak inducer of apoptosis induces inflammation in Graves' orbital fibroblasts. PLoS One 2018; 13:e0209583. [PMID: 30576385 PMCID: PMC6303076 DOI: 10.1371/journal.pone.0209583] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023] Open
Abstract
Tumor necrosis factor-like weak inducer of apoptosis (TWEAK), along with its receptor fibroblast growth factor-inducible (Fn)14, is associated with various biological activities including inflammation. However, its role in the pathogenesis of Graves’ orbitopathy (GO) is unknown. In this study, we investigated the mechanism by which TWEAK regulates inflammatory signaling in orbital fibroblasts from GO patients. We found that TWEAK and tumor necrosis factor-α (TNFA) mRNA levels were upregulated in GO as compared to non-GO tissue samples. TWEAK, TNF receptor (TNFR)1, TNFR2, and TNFR superfamily member 12A mRNA, and TWEAK and Fn14 protein levels were increased by interleukin (IL)-1β and TNF-α treatment. Treatment with exogenous recombinant TWEAK increased the transcript and protein expression of the pro-inflammatory cytokines IL-6, IL-8, and monocyte chemoattractant protein-1 to a greater extent in GO than in non-GO cells, while treatment with the anti-Fn14 antibody ITEM4 suppressed TWEAK-induced pro-inflammatory cytokine release and hyaluronan production. Additionally, the serum level of TWEAK was higher in Graves’ disease patients with (341.86 ± 86.3 pg/ml) as compared to those without (294.09 ± 41.44 pg/ml) GO and healthy subjects (255.33 ± 39.38 pg/ml), and was positively correlated with clinical activity score (r = 0.629, P < 0.001) and thyroid binding immunoglobulin level (r = 0.659, P < 0.001). These results demonstrate that TWEAK/Fn14 signaling contributes to GO pathogenesis. Moreover, serum TWEAK level is a potential diagnostic biomarker for inflammatory GO, and modulating TWEAK activity may be an effective therapeutic strategy for suppressing inflammation and tissue remodeling in GO.
Collapse
Affiliation(s)
- Sung Jun Lee
- Yonsei Bon Eye Clinic, Seoul, Korea
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jinjoo Kim
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - JaeSang Ko
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Jig Lee
- Department of Endocrinology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyoung Jun Koh
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Sook Yoon
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
36
|
Xu RD, Feng F, Yu XS, Liu ZD, Lao LF. miR-149-5p inhibits cell growth by regulating TWEAK/Fn14/PI3K/AKT pathway and predicts favorable survival in human osteosarcoma. Int J Immunopathol Pharmacol 2018; 32:2058738418786656. [PMID: 30014744 PMCID: PMC6050806 DOI: 10.1177/2058738418786656] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) as small non-coding RNAs act as either tumor suppressors or
oncogenes in human cancers, of which miR-149-5p (miR-149) is involved in tumor
growth and metastasis, but its role and molecular mechanisms underlying
osteosarcoma growth are poorly understood. The correlation of miR-149 expression
with clinicopathological characteristics and prognosis in patients with sarcoma
was analyzed by The Cancer Genome Atlas (TCGA) RNA-sequencing data. Osteosarcoma
cell growth affected by miR-149 was evaluated by
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and colony
formation assays. As a result, we found that the expression level of miR-149 was
markedly downregulated in human sarcoma samples and were negatively associated
with tumor size, acting as an independent prognostic factor for overall survival
of the sarcoma patients. Restoration of miR-149 expression suppressed
osteosarcoma cell growth, while its knockdown reversed these effects.
Furthermore, we identified TNFRSF12A (TNF receptor superfamily member 12A), also
called fibroblast growth factor–inducible 14 (Fn14) as a direct target of
miR-149, and TNFRSF12A and its ligand TNFSF12 (TNF superfamily member 12), also
called tumor necrosis factor–related weak inducer of apoptosis (TWEAK), were
both negatively correlated with miR-149 expression in sarcoma samples. Knockdown
of TNFRSF12A suppressed cell growth, but its overexpression weakened the
antiproliferative effects of miR-149 via the PI3K/AKT (AKT serine/threonine
kinase) signaling pathway. Altogether, our findings show that miR-149 functions
as a tumor suppressor in osteosarcoma via inhibition of the TWEAK–Fn14 axis and
represents a potential therapeutic target in patients with osteosarcoma.
Collapse
Affiliation(s)
- Rui-Da Xu
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Feng
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Sheng Yu
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zu-De Liu
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Feng Lao
- Department of Orthopaedic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
37
|
Watanabe M, Toyomura T, Wake H, Liu K, Teshigawara K, Takahashi H, Nishibori M, Mori S. The C-terminal region of tumor necrosis factor like weak inducer of apoptosis is required for interaction with advanced glycation end products. Biotechnol Appl Biochem 2018; 66:254-260. [PMID: 30403295 DOI: 10.1002/bab.1706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 11/01/2018] [Indexed: 12/21/2022]
Abstract
Previously, we found that endogenously produced pro-inflammatory molecules, advanced glycation end products (AGEs), interact with tumor necrosis factor-like weak inducer of apoptosis (TWEAK), and attenuate its immunomodulatory function. In the present study, to elucidate the mechanism by which AGEs attenuate TWEAK function, we searched for regions responsible for TWEAK-AGE interaction using TWEAK deletion mutants. Pull-down assays with the TWEAK mutants and AGEs revealed that the C-terminal half of TWEAK, which is the region essential for receptor stimulation, was required for this interaction. On the other hand, the N-terminal deletion mutants did not exhibit a significant decrease in AGE binding. Moreover, a moderate decrease in the AGE binding by double-deletion in quartered C-terminal half regions and a substantial decrease by triple-deletion in this region were observed. In addition, full-length TWEAK stimulated IL-8 gene expression in endothelial EA.hy.926 cells, whereas the triple-deletion mutant lost much of this activity, suggesting that the TWEAK-AGE interaction sites overlap with the region needed to exert normal function of TWEAK. Our present findings may help to elucidate the pathophysiological roles of the TWEAK-AGE interaction for prevention and treatment of AGE-related inflammatory diseases.
Collapse
Affiliation(s)
- Masahiro Watanabe
- Department of Pharmacology, School of Pharmacy, Shujitsu University, Okayama, Japan
| | - Takao Toyomura
- Department of Pharmacology, School of Pharmacy, Shujitsu University, Okayama, Japan
| | - Hidenori Wake
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Keyue Liu
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Kiyoshi Teshigawara
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hideo Takahashi
- Department of Pharmacology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Masahiro Nishibori
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Shuji Mori
- Department of Pharmacology, School of Pharmacy, Shujitsu University, Okayama, Japan
| |
Collapse
|
38
|
Soluble TNF-like weak inducer of apoptosis (TWEAK) enhances poly(I:C)-induced RIPK1-mediated necroptosis. Cell Death Dis 2018; 9:1084. [PMID: 30349023 PMCID: PMC6197222 DOI: 10.1038/s41419-018-1137-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 10/03/2018] [Accepted: 10/08/2018] [Indexed: 12/14/2022]
Abstract
TNF-like weak inducer of apoptosis (TWEAK) and inhibition of protein synthesis with cycloheximide (CHX) sensitize for poly(I:C)-induced cell death. Notably, although CHX preferentially enhanced poly(I:C)-induced apoptosis, TWEAK enhanced primarily poly(I:C)-induced necroptosis. Both sensitizers of poly(I:C)-induced cell death, however, showed no major effect on proinflammatory poly(I:C) signaling. Analysis of a panel of HeLa-RIPK3 variants lacking TRADD, RIPK1, FADD, or caspase-8 expression revealed furthermore similarities and differences in the way how poly(I:C)/TWEAK, TNF, and TRAIL utilize these molecules for signaling. RIPK1 turned out to be essential for poly(I:C)/TWEAK-induced caspase-8-mediated apoptosis but was dispensable for this response in TNF and TRAIL signaling. TRADD-RIPK1-double deficiency differentially affected poly(I:C)-triggered gene induction but abrogated gene induction by TNF completely. FADD deficiency abrogated TRAIL- but not TNF- and poly(I:C)-induced necroptosis, whereas TRADD elicited protective activity against all three death inducers. A general protective activity against poly(I:C)-, TRAIL-, and TNF-induced cell death was also observed in FLIPL and FLIPS transfectrants.
Collapse
|
39
|
Autophagy differentially regulates TNF receptor Fn14 by distinct mammalian Atg8 proteins. Nat Commun 2018; 9:3744. [PMID: 30218067 PMCID: PMC6138730 DOI: 10.1038/s41467-018-06275-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 07/26/2018] [Indexed: 02/07/2023] Open
Abstract
Autophagy, a conserved membrane trafficking process, sequesters cytoplasmic components into autophagosomes and targets them for lysosomal degradation. The TNF receptor Fn14 participates in multiple intracellular signaling pathways and is strongly induced upon tissue injury and solid tumorigenesis. While Fn14 is a short-lived protein, the regulation of its levels is largely obscure. Here we uncover a role for autophagy in Fn14 turnover, wherein specific core autophagy Atg8 proteins play distinct roles: Fn14 accumulates in the ERGIC in absence of GABARAP but within endosomes in the vicinity of autophagic membranes in absence of GATE-16. Moreover, GABARAP regulates overall cellular levels of Fn14, whereas GATE-16 regulates TWEAK signaling by Fn14 and thereby NF-κB activity. These findings not only implicate different Atg8 proteins in distinct roles within the mechanism of selective autophagic regulation of Fn14, but may also provide a more general view of their role in mediating autophagosome biogenesis from different membrane sources.
Collapse
|
40
|
Differential Roles of Tumor Necrosis Factor Ligand Superfamily Members as Biomarkers in Pancreatic Cancer. J Clin Med 2018; 7:jcm7070175. [PMID: 30011858 PMCID: PMC6068811 DOI: 10.3390/jcm7070175] [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: 06/18/2018] [Revised: 07/04/2018] [Accepted: 07/12/2018] [Indexed: 12/18/2022] Open
Abstract
The tumor necrosis factor⁻related weak inducer of apoptosis (TWEAK) belongs to the tumor necrosis factor ligand superfamily, which was shown to play an important role in inflammatory and malignant gastrointestinal diseases, including colitis or colorectal cancer. However, in contrast to other members of the TNF ligand superfamily, its role as a biomarker in pancreatic cancer is currently unknown. We analyzed serum levels of A proliferation-inducing ligand (APRIL) and TWEAK in 134 patients with pancreatic cancer. Results were compared with 50 healthy controls and correlated with clinical data. Intratumoral expression of APRIL and TWEAK in pancreatic cancer was analysed using the datasets made available by the TCGA-LIHC project. APRIL serum levels were significantly elevated in patients with pancreatic cancer compared to healthy controls, which is in line with previous findings. Notably, the diagnostic accuracy of circulating APRIL levels was similar to CA19-9, an established tumor marker for pancreatic cancer. In contrast, serum concentrations of TWEAK were decreased in pancreatic cancer patients. Interestingly, no differences in TWEAK concentrations became apparent between different clinical subgroups of pancreatic cancer. Moreover, within our cohort of patients, TWEAK levels did not correlate with the patients' prognosis and the diagnostic as well as prognostic potential of TWEAK was lower than CA 19-9, when analyzed in this setting. Finally, using data from the TCGA-LIHC project, we demonstrate that expression levels of TWEAK and APRIL represent prognostic markers for patients' survival according to Kaplan-Meier curve analyses. TWEAK and APRIL serum concentrations are regulated differently in patients with pancreatic cancer, highlighting diverse roles of variant TNF ligands in this type of cancer.
Collapse
|
41
|
FN14 Blockade on Pulmonary Microvascular Endothelial Cells Improves the Outcome of Sepsis-Induced Acute Lung Injury. Shock 2018; 49:213-220. [DOI: 10.1097/shk.0000000000000915] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
42
|
Guan F, Wang L, Hao S, Wu Z, Bai J, Kang Z, Zhou Q, Chang H, Yin H, Li D, Tian K, Ma J, Zhang G, Zhang J. Retinol dehydrogenase-10 promotes development and progression of human glioma via the TWEAK-NF-κB axis. Oncotarget 2017; 8:105262-105275. [PMID: 29285249 PMCID: PMC5739636 DOI: 10.18632/oncotarget.22166] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 09/25/2017] [Indexed: 12/03/2022] Open
Abstract
Retinol dehydrogenase-10 (RDH10) is a member of the short-chain dehydrogenase/reductase family, which plays an important role in retinoic acid (RA) synthesis. Here, we show that RDH10 is highly expressed in human gliomas, and its expression correlates with tumor grade and patient survival times. In vitro, lentivirus-mediated shRNA knockdown of RDH10 suppressed glioma cell proliferation, survival, and invasiveness and cell cycle progression. In vivo, RDH10 knockdown reduced glioma growth in nude mice. Microarray analysis revealed that RDH10 silencing reduces expression of TNFRSF12A (Fn14), TNFSF12 (TWEAK), TRAF3, IKBKB (IKK-β), and BMPR2, while it increases expression of TRAF1, NFKBIA (IκBα), NFKBIE (IκBε), and TNFAIP3. This suggests that RDH10 promotes glioma cell proliferation and survival by regulating the TWEAK-NF-κB axis, and that it could potentially serve as a novel target for human glioma treatment.
Collapse
Affiliation(s)
- Feng Guan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuyu Hao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jian Bai
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Zhuang Kang
- Department of Glioma, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Quan Zhou
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hong Chang
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hui Yin
- Department of Social Medicine and Health Education, School of Public Health, Peking University, Beijing, China
| | - Da Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kaibin Tian
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Junpeng Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guijun Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Junting Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
43
|
Liu Q, Xiao S, Xia Y. TWEAK/Fn14 Activation Participates in Skin Inflammation. Mediators Inflamm 2017; 2017:6746870. [PMID: 29038621 PMCID: PMC5606047 DOI: 10.1155/2017/6746870] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/01/2017] [Indexed: 02/07/2023] Open
Abstract
Tumor necrosis factor- (TNF-) like weak inducer of apoptosis (TWEAK) participates in multiple biological activities via binding to its sole receptor-fibroblast growth factor-inducible 14 (Fn14). The TWEAK/Fn14 signaling pathway is activated in skin inflammation and modulates the inflammatory responses of keratinocytes by activating nuclear factor-κB signals and enhancing the production of several cytokines, including interleukins, monocyte chemotactic protein-1, RANTES (regulated on activation, normal T cell expressed and secreted), and interferon gamma-induced protein 10. Mild or transient TWEAK/Fn14 activation contributes to tissular repair and regeneration while excessive or persistent TWEAK/Fn14 signals may lead to severe inflammatory infiltration and tissue damage. TWEAK also regulates cell fate of keratinocytes, involving the function of Fn14-TNF receptor-associated factor-TNF receptor axis. By recruiting inflammatory cells, promoting cytokine production, and regulating cell fate, TWEAK/Fn14 activation plays a pivotal role in the pathogenesis of various skin disorders, such as psoriasis, atopic dermatitis, cutaneous vasculitis, human papillomavirus infection and related skin tumors, and cutaneous autoimmune diseases. Therefore, the TWEAK/Fn14 pathway may be a potential target for the development of novel therapeutics for skin inflammatory diseases.
Collapse
Affiliation(s)
- Qilu Liu
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Shengxiang Xiao
- 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
|
44
|
Prigozhina TB, Szafer F, Aronin A, Tzdaka K, Amsili S, Makdasi E, Shani N, Dranitzki Elhalel M. Fn14·TRAIL fusion protein is oligomerized by TWEAK into a superefficient TRAIL analog. Cancer Lett 2017; 400:99-109. [DOI: 10.1016/j.canlet.2017.04.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 04/12/2017] [Accepted: 04/14/2017] [Indexed: 01/08/2023]
|
45
|
TWEAK/Fn14 Activation Contributes to the Pathogenesis of Bullous Pemphigoid. J Invest Dermatol 2017; 137:1512-1522. [DOI: 10.1016/j.jid.2017.03.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/06/2017] [Accepted: 03/07/2017] [Indexed: 01/05/2023]
|
46
|
Watanabe M, Toyomura T, Wake H, Liu K, Teshigawara K, Takahashi H, Nishibori M, Mori S. Advanced glycation end products attenuate the function of tumor necrosis factor-like weak inducer of apoptosis to regulate the inflammatory response. Mol Cell Biochem 2017; 434:153-162. [DOI: 10.1007/s11010-017-3045-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/25/2017] [Indexed: 12/31/2022]
|
47
|
González-Sánchez DA, Álvarez CM, Vásquez G, Gómez-Puerta JA. Papel de la vía de señalización del TWEAK/Fn14 en la nefritis lúpica y otros escenarios clínicos. Nefrologia 2017; 37:118-125. [DOI: 10.1016/j.nefro.2016.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 05/11/2016] [Accepted: 05/12/2016] [Indexed: 02/06/2023] Open
|
48
|
Min JK. Is Urinary Tumor Necrosis Factor-like Weak Inducer of Apoptosis a Biomarker of Lupus Nephritis? JOURNAL OF RHEUMATIC DISEASES 2017. [DOI: 10.4078/jrd.2017.24.3.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Jun-Ki Min
- Division of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Bucheon St. Mary's Hospital, Bucheon, Korea
| |
Collapse
|
49
|
Armstrong CL, Galisteo R, Brown SA, Winkles JA. TWEAK activation of the non-canonical NF-κB signaling pathway differentially regulates melanoma and prostate cancer cell invasion. Oncotarget 2016; 7:81474-81492. [PMID: 27821799 PMCID: PMC5348407 DOI: 10.18632/oncotarget.13034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/14/2016] [Indexed: 12/22/2022] Open
Abstract
Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a multifunctional cytokine that binds with high affinity to a plasma membrane-anchored receptor named Fn14. Both TWEAK and Fn14 expression has been detected in human cancer tissue, and studies have shown that TWEAK/Fn14 signaling can promote either "pro-cancer" or "anti-cancer" cellular effects in vitro, depending on the cancer cell line under investigation. In this study, we engineered murine B16 melanoma cells to secrete high levels of soluble TWEAK and examined their properties. TWEAK production by B16 cells preferentially activated the non-canonical NF-κB signaling pathway and increased the expression of several previously described TWEAK-inducible genes, including Fn14. TWEAK overexpression in B16 cells inhibited both cell growth and invasion in vitro. The TWEAK-mediated reduction in B16 cell invasive capacity was dependent on activation of the non-canonical NF-κB signaling pathway. Finally, we found that this same signaling pathway was also important for TWEAK-stimulated human DU145 prostate cancer cell invasion. Therefore, even though TWEAK:Fn14 binding activates non-canonical NF-κB signaling in both melanoma and prostate cancer cells, this shared cellular response can trigger a very different downstream outcome (inhibition or stimulation of cell invasiveness, respectively).
Collapse
Affiliation(s)
- Cheryl L. Armstrong
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rebeca Galisteo
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sharron A.N. Brown
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jeffrey A. Winkles
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
50
|
Abstract
INTRODUCTION Preeclampsia is a major pregnancy disease, explained partly by genetic predispositions. STOX1, a transcription factor discovered in 2005, was the first gene directly associated with genetic forms of the disease. Alterations of STOX1 expression as well as STOX1 variants have also been associated to Alzheimer's disease. These observations make of this gene a putative therapeutic target. Area covered: Two major isoforms (STOX1A and STOX1B) are encoded by the gene and are theoretically able to compete for the same binding site, while only the most complete (STOX1A) is supposed to be able to activate gene expression. This makes the ratio between STOX1A and STOX1B as well as their position inside the cell (nucleus or cytoplasm) crucial to understand how STOX1 functions. STOX1 appears to have multiple gene targets, especially in pathways connected to inflammation, oxidative stress, and cell cycle. Expert opinion: STOX1-directed therapies, could be directed either towards its targets (genes or pathways), or directly at STOX1. For this the addressing of STOX1 to various cell compartments could theoretically be modified; also it could be possible of altering the balance between the two isoforms, through selectively inhibiting one of them, possibly improving the outcomes in severe preeclampsia.
Collapse
Affiliation(s)
- Daniel Vaiman
- a Department of Development, Reproduction and Cancer , Institut Cochin , Paris , France
| | - Francisco Miralles
- a Department of Development, Reproduction and Cancer , Institut Cochin , Paris , France
| |
Collapse
|