1
|
Du W, Ren N, Xu Y, Chen X. Programmed cell death 4 governs NLRP3-mediated pyroptosis in septic lung disorders. Mol Biol Rep 2024; 51:77. [PMID: 38183433 DOI: 10.1007/s11033-023-08948-7] [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: 07/19/2023] [Accepted: 11/21/2023] [Indexed: 01/08/2024]
Abstract
INTRODUCTION Sepsis is a pathogenic syndrome of prolonged excessive inflammation and immunosuppression produced by invading pathogens. Programmed cell death 4 (PDCD4) may be implicated in a range of inflammatory lesions, and this study aimed to confirm the involvement of PDCD4 in septic lung injury. MATERIALS AND METHODS Mice and bronchial epithelial 16HBE cells were separately subjected to CLP and LPS to generate in vivo and in vitro models. Following the level of PDCD4 was determined, the impacts of PDCD4 knockdown on mouse lung injury degree, inflammation, apoptosis, and pyroptosis levels were evaluated. Afterward, cells were treated with the NLRP3 agonist, and the influences of NLRP3 activation on the regulations of PDCD4 knockdown were determined. RESULTS PDCD4 was elevated following mice developed septic lung injury, PDCD4 knockdown ameliorated septic lung injury and reduced lung inflammation and apoptosis. Moreover, PDCD4 knockdown suppressed NLRP3-mediated pyroptosis, indicating that PDCD4 also mediated pyroptosis. According to cellular models, NLRP3 activation broke the effects of PDCD4 knockdown on cells. CONCLUSIONS The current study reveals that PDCD4 governs NLRP3-mediated pyroptosis in septic lung injury. PDCD4 is not only related to apoptosis and expands the knowledge of PDCD4 regulation of different cell death modes.
Collapse
Affiliation(s)
- Wenjie Du
- Department of Emergency Internal Medicine, the Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Qingdao, 266000, Shandong, China
| | - Na Ren
- Department of Emergency Internal Medicine, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao, 266033, Shandong, China
| | - Yan Xu
- Quality Control Department, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao, 266033, Shandong, China
| | - Xiao Chen
- Department of Emergency Internal Medicine, the Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Qingdao, 266000, Shandong, China.
| |
Collapse
|
2
|
Montero JC, Del Carmen S, Abad M, Sayagués JM, Barbáchano A, Fernández-Barral A, Muñoz A, Pandiella A. An amino acid transporter subunit as an antibody-drug conjugate target in colorectal cancer. J Exp Clin Cancer Res 2023; 42:200. [PMID: 37559159 PMCID: PMC10410906 DOI: 10.1186/s13046-023-02784-0] [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: 03/10/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Advanced colorectal cancer (CRC) is difficult to treat. For that reason, the development of novel therapeutics is necessary. Here we describe a potentially actionable plasma membrane target, the amino acid transporter protein subunit CD98hc. METHODS Western blot and immunohistochemical analyses of CD98hc protein expression were carried out on paired normal and tumoral tissues from patients with CRC. Immunofluorescence and western studies were used to characterize the action of a DM1-based CD98hc-directed antibody-drug conjugate (ADC). MTT and Annexin V studies were performed to evaluate the effect of the anti-CD98hc-ADC on cell proliferation and apoptosis. CRISPR/Cas9 and shRNA were used to explore the specificity of the ADC. In vitro analyses of the antitumoral activity of the anti-CD98hc-ADC on 3D patient-derived normal as well as tumoral organoids were also carried out. Xenografted CRC cells and a PDX were used to analyze the antitumoral properties of the anti-CD98hc-ADC. RESULTS Genomic as well proteomic analyses of paired normal and tumoral samples showed that CD98hc expression was significantly higher in tumoral tissues as compared to levels of CD98hc present in the normal colonic tissue. In human CRC cell lines, an ADC that recognized the CD98hc ectodomain, reached the lysosomes and exerted potent antitumoral activity. The specificity of the CD98hc-directed ADC was demonstrated using CRC cells in which CD98hc was decreased by shRNA or deleted using CRISPR/Cas9. Studies in patient-derived organoids verified the antitumoral action of the anti-CD98hc-ADC, which largely spared normal tissue-derived colon organoids. In vivo studies using xenografted CRC cells or patient-derived xenografts confirmed the antitumoral activity of the anti-CD98hc-ADC. CONCLUSIONS The studies herewith reported indicate that CD98hc may represent a novel ADC target that, upon well-designed clinical trials, could be used to increase the therapeutic armamentarium against CRC.
Collapse
Affiliation(s)
- Juan Carlos Montero
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca), Salamanca, Spain.
- Department of Pathology and IBSAL, University Hospital of Salamanca, Salamanca, Spain.
- CIBERONC, Madrid, Spain.
| | - Sofía Del Carmen
- Department of Pathology and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - Mar Abad
- Department of Pathology and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - José M Sayagués
- Department of Pathology and IBSAL, University Hospital of Salamanca, Salamanca, Spain
- CIBERONC, Madrid, Spain
| | - Antonio Barbáchano
- CIBERONC, Madrid, Spain
- Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-Autonomous University of Madrid, and Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain
| | - Asunción Fernández-Barral
- CIBERONC, Madrid, Spain
- Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-Autonomous University of Madrid, and Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain
| | - Alberto Muñoz
- CIBERONC, Madrid, Spain
- Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-Autonomous University of Madrid, and Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain
| | - Atanasio Pandiella
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca), Salamanca, Spain.
- CIBERONC, Madrid, Spain.
| |
Collapse
|
3
|
Neuregulin modulates hormone receptor levels in breast cancer through concerted action on multiple signaling pathways. Clin Sci (Lond) 2023; 137:1-15. [PMID: 36511917 PMCID: PMC9805957 DOI: 10.1042/cs20220472] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/05/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022]
Abstract
The Neuregulins (NRGs) are growth factors that bind and activate ErbB/HER receptor tyrosine kinases. Some reports have described an interplay between this ligand-receptor system and hormonal receptors in breast cancer. However, the mechanisms by which NRGs regulate hormonal receptor signaling have not been sufficiently described. Here, we show that in breast cancer cells the activation of NRG receptors down-regulated ERα through a double mechanism that included post-transcriptional and transcriptional effects. This regulation required the concerted participation of three signaling routes: the PI3K/AKT/mTOR, ERK1/2, and ERK5 pathways. Moreover, these three routes were also involved in the phosphorylation of ERα at serines 118 and 167, two residues implicated in resistance to endocrine therapies. On the other hand, NRGs conferred resistance to fulvestrant in breast cancer cells and this resistance could be reversed when the three pathways activated by NRGs were simultaneously inhibited. Our results indicate that estrogen receptor-positive (ER+) breast tumors that can have access to NRGs may be resistant to fulvestrant. This resistance could be overcome if strategies to target the three main pathways involved in the interplay between NRG receptors and ERα could be developed.
Collapse
|
4
|
Materials, workflows and applications of IMAC for phosphoproteome profiling in the recent decade: A review. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
5
|
Qiu J, Jiang T, Yang G, Gong Y, Zhang W, Zheng X, Chen H, Hong Z. Neratinib Exerts Dual Effects on Cartilage Degradation and Osteoclast Production in Osteoarthritis by Inhibiting the Activation of the MAPK/NF-κB Signaling Pathways. Biochem Pharmacol 2022; 205:115155. [PMID: 35820500 DOI: 10.1016/j.bcp.2022.115155] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/04/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022]
Abstract
Osteoarthritis (OA) is a degenerative disease caused by the progressive destruction of cartilage and subchondral bone. [1] Studies have shown that by inhibiting the degradation of cartilage cells and the loss of subchondral bone, OA can be prevented and treated. Neratinib, as a small molecule compound with anti-inflammatory and anti-tumor properties, is a very effective inhibitor of IL-1β-induced chondrocyte inflammation and anabolic metabolism. By investigating the effect of neratinib in ATDC5 chondrocytes, the study finds that neratinib reduces inflammation by inhibiting the MAPK and NF-κB signaling pathways, and at the same time reduces pyrolysis (indicated by the results of reverse transcription quantitative PCR and western blotting). For anabolic metabolism, after high-density cell culture, IL-1β-induced catalytic changes and degradation of the extracellular matrix were evaluated by toluidine blue staining. Since osteoclasts are key participants in the process of subchondral bone remodeling in OA, we also studied the effect of neratinib on the maturation of osteoclasts. The results showed that neratinib also acts as an anti-osteoclast agent in vitro. By inhibiting the NF-κB and MAPK pathways, it reduces the expression of osteoclast-related genes, thereby inhibiting RANKL-induced osteoclastogenesis. The results of in vivo animal experiments supported the conclusions from the experiments in vitro. Neratinib inhibited both the destruction of medial meniscus induced cartilage degradation and osteoclast formation, which proves that neratinib has a dual effect, protecting cartilage and inhibiting osteoclast formation. These results indicate that neratinib can be a brand-new latent strategy for the treatment of OA.
Collapse
Affiliation(s)
- Jianxin Qiu
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China; Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
| | - Ting Jiang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China; Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
| | - Guangyong Yang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China; Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
| | - Yuhang Gong
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China; Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
| | - Weikang Zhang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China; Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
| | - Xiaohang Zheng
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China; Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
| | - Haixiao Chen
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China; Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China.
| | - Zhenghua Hong
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China; Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China.
| |
Collapse
|
6
|
Cai Q, Yang HS, Li YC, Zhu J. Dissecting the Roles of PDCD4 in Breast Cancer. Front Oncol 2022; 12:855807. [PMID: 35795053 PMCID: PMC9251513 DOI: 10.3389/fonc.2022.855807] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 05/12/2022] [Indexed: 11/29/2022] Open
Abstract
The human programmed cell death 4 (PDCD4) gene was mapped at chromosome 10q24 and encodes the PDCD4 protein comprised of 469 amino acids. PDCD4 inhibits protein translation PDCD4 inhibits protein translation to suppress tumor progression, and its expression is frequently decreased in breast cancer. PDCD4 blocks translation initiation complex by binding eIF4A via MA-3 domains or by directly binding 5’ mRNA internal ribosome entry sites with an RNA binding domain to suppress breast cancer progression and proliferation. Numerous regulators and biological processes including non-coding RNAs, proteasomes, estrogen, natural compounds and inflammation control PDCD4 expression in breast cancer. Loss of PDCD4 expression is also responsible for drug resistance in breast cancer. HER2 activation downregulates PDCD4 expression by activating MAPK, AKT, and miR-21 in aromatase inhibitor-resistant breast cancer cells. Moreover, modulating the microRNA/PDCD4 axis maybe an effective strategy for overcoming chemoresistance in breast cancer. Down-regulation of PDCD4 is significantly associated with short overall survival of patients, which suggests that PDCD4 may be an independent prognostic marker for breast cancer.
Collapse
Affiliation(s)
- Qian Cai
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovasular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Hsin-Sheng Yang
- Department of Toxicology and Cancer Biology, Collage of Medicine, University of Kentucky, Lexington, KY, United States
| | - Yi-Chen Li
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Jiang Zhu
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, China
- *Correspondence: Jiang Zhu,
| |
Collapse
|
7
|
Montero JC, Calvo-Jiménez E, Del Carmen S, Abad M, Ocaña A, Pandiella A. Surfaceome analyses uncover CD98hc as an antibody drug-conjugate target in triple negative breast cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:106. [PMID: 35317825 PMCID: PMC8941813 DOI: 10.1186/s13046-022-02330-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/12/2022] [Indexed: 11/10/2022]
Abstract
Background Despite the incorporation of novel therapeutics, advanced triple negative breast cancer (TNBC) still represents a relevant clinical problem. Considering this, as well as the clinical efficacy of antibody-drug conjugates (ADCs), we aimed at identifying novel ADC targets that could be used to treat TNBC. Methods Transcriptomic analyses were performed on TNBC and normal samples from three different studies. Plasma membrane proteins of three cell lines representative of the TNBC subtype were identified by cell surface biotinylation or plasma membrane isolation, followed by analyses of cell surface proteins using the Surfaceome online tool. Immunofluorescence and western studies were used to characterize the action of a CD98hc-directed ADC, which was prepared by in house coupling of emtansine to an antibody that recognized the ectodomain of CD98hc. Xenografted TNBC cells were used to analyze the antitumoral properties of the anti-CD98hc ADC. Results Comparative genomic studies between normal breast and TNBC tissues, together with proteomic and bioinformatic analyses resulted in the elaboration of a catalog of potential ADC targets. One of them, the CD98hc transmembrane protein, was validated as an ADC target. An antibody recognizing the ectodomain of CD98hc efficiently internalized and reached the lysosomal compartment. An emtansine-based ADC derived from such antibody was prepared and showed antitumoral properties in TNBC in vitro and in vivo models. Mechanistically, the anti-CD98hc ADC blocked cell cycle progression, that was followed by cell death caused by mitotic catastrophe. Conclusions This work describes a list of potential ADC targets in TNBC and validates one of them, the transmembrane protein CD98hc. The studies presented here also demonstrate the robustness of the multiomic approach herewith described to identify novel potential ADC targets. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02330-4.
Collapse
Affiliation(s)
- Juan Carlos Montero
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Salamanca, Spain. .,Department of Pathology and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007, Salamanca, Spain.
| | - Elisa Calvo-Jiménez
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Salamanca, Spain
| | - Sofía Del Carmen
- Department of Pathology and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007, Salamanca, Spain
| | - Mar Abad
- Department of Pathology and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007, Salamanca, Spain
| | | | - Atanasio Pandiella
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Salamanca, Spain
| |
Collapse
|