1
|
Cavalcanti L, Francati S, Ferraguti G, Fanfarillo F, Peluso D, Barbato C, Greco A, Minni A, Petrella C. Lipocalin-2, Matrix Metalloproteinase-9, and MMP-9/NGAL Complex in Upper Aerodigestive Tract Carcinomas: A Pilot Study. Cells 2025; 14:506. [PMID: 40214460 PMCID: PMC11988122 DOI: 10.3390/cells14070506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
Upper aerodigestive tract (UADT) carcinomas have a high and rapidly increasing incidence, particularly in industrialized countries. The identification of diagnostic and prognostic biomarkers remains a key objective in oncological research. However, conflicting data have been reported regarding Lipocalin-2 (LCN-2 or NGAL), Matrix Metalloproteinase-9 (MMP-9), and the MMP-9/NGAL complex in UADT carcinomas. For this reason, the primary aim of this study was to investigate the involvement and modulation of the LCN-2 system in UADT cancer by selecting patients at first diagnosis and excluding any pharmacological or interventional treatments that could act as confounding factors. In this clinical retrospective pilot study, we investigated LCN-2 and MMP-9 tissue gene expression, as well as circulating levels of LCN-2, MMP-9, and the MMP-9/NGAL complex. Our findings revealed a downregulation of LCN-2 and an upregulation of MMP-9 gene expression in tumor tissues compared to healthy counterparts. A similar trend was observed in circulating levels, with decreased LCN-2 and increased MMP-9 in cancer patients compared to healthy controls. Additionally, serum levels of the MMP-9/NGAL complex were significantly elevated in UADT cancer patients relative to controls. Our study suggests a potentially distinct role for the free form of LCN-2 and its conjugated form (MMP-9/NGAL complex) in UADT tumors. These findings not only provide new insights into the molecular mechanisms underlying tumor progression but also highlight the potential clinical relevance of these biomarkers. The differential expression patterns observed suggest that the LCN-2 and MMP-9/NGAL complex could serve as valuable tools for improving early diagnosis, monitoring disease progression, and potentially guiding therapeutic strategies. Further research is needed to validate their utility in clinical settings and to explore their prognostic and predictive value in personalized treatment approaches.
Collapse
Affiliation(s)
- Luca Cavalcanti
- Department of Sensory Organs, Sapienza University of Rome, 00161 Roma, Italy; (L.C.); (A.G.); (A.M.)
| | - Silvia Francati
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Roma, Italy; (S.F.); (G.F.); (F.F.)
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Roma, Italy; (S.F.); (G.F.); (F.F.)
| | - Francesca Fanfarillo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Roma, Italy; (S.F.); (G.F.); (F.F.)
| | - Daniele Peluso
- PhD School of Applied Medical-Surgical Sciences, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Roma, Italy;
- Department of Biology, University of Rome “Tor Vergata”, 00133 Roma, Italy
| | - Christian Barbato
- Institute of Biochemistry and Cell Biology (IBBC-CNR), 00161 Roma, Italy;
| | - Antonio Greco
- Department of Sensory Organs, Sapienza University of Rome, 00161 Roma, Italy; (L.C.); (A.G.); (A.M.)
| | - Antonio Minni
- Department of Sensory Organs, Sapienza University of Rome, 00161 Roma, Italy; (L.C.); (A.G.); (A.M.)
- Division of Otolaryngology-Head and Neck Surgery, San Camillo de Lellis Hospital, ASL Rieti-Sapienza University, Viale Kennedy, 02100 Rieti, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology (IBBC-CNR), 00161 Roma, Italy;
| |
Collapse
|
2
|
Dave S, Patel B. The lipocalin saga: Insights into its role in cancer-associated cachexia. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167684. [PMID: 39837432 DOI: 10.1016/j.bbadis.2025.167684] [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: 08/07/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
Cancer-associated cachexia (CAC) is a debilitating condition, observed in patients with advanced stages of cancer. It is marked by ongoing weight loss, weakness, and nutritional impairment. Lower tolerance of chemotherapeutic agents and radiation therapy makes it difficult to treat CAC. Anorexia is a significant contributor to worsening CAC. Anorexia can be found in the early or advanced stages of cancer. Anorexia in cancer patients arises from a confluence of factors. Tumor-related inflammatory cytokines can directly impact the gastrointestinal tract, leading to dysphagia and compromised gut function. Additionally, increased serotonin and hormonal disruptions lead to early satiety, suppressing appetite. Due to the complexities in the pathogenesis of the disease, identifying druggable targets is a challenge. Research is ongoing to identify novel targets for the treatment of this condition. Recent research suggests a potential link between elevated levels of Lipocalin 2 (LCN2) and cachexia in cancer patients. LCN2, a glycoprotein primarily released by neutrophils, is implicated in numerous illnesses, including skin disorders, cancer, atherosclerosis, and type 2 diabetes. LCN2 suppresses hunger by binding to the melanocortin-4 receptors. Several in vitro, in vivo, and clinical studies indicate the association between LCN2 levels and appetite suppression. Further research should be explored emphasizing the significance of well-crafted clinical trials to confirm LCN2's usefulness as a therapeutic target and its ability to help cancer patients who are suffering from the fatal hallmark of cachexia. This review explores LCN2's function in the multifaceted dynamics of CAC and anorexia.
Collapse
Affiliation(s)
- Srusti Dave
- National Forensic Sciences University, Gandhinagar 382007, Gujarat, India
| | - Bhoomika Patel
- National Forensic Sciences University, Gandhinagar 382007, Gujarat, India.
| |
Collapse
|
3
|
HUANG BAOXING, JIA ZICHANG, FU CHENCHEN, CHEN MOXIAN, SU ZEZHUO, CHEN YUNSHENG. Oncogenic and tumor-suppressive roles of Lipocalin 2 (LCN2) in tumor progression. Oncol Res 2025; 33:567-575. [PMID: 40109857 PMCID: PMC11915076 DOI: 10.32604/or.2024.051672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/17/2024] [Indexed: 03/22/2025] Open
Abstract
Lipocalin-2 (LCN2) is a member of the lipocalin superfamily with multiple functions and can participate in the transport of a variety of small lipophilic ligands in vivo. LCN2 is significantly expressed in various tumors and plays an important role in regulating tumor cell proliferation, invasion, and metastasis. The specific actions of LCN2 in tumors may vary depending on the particular type of cancer involved. In this review, we provide an extensive overview of the transcriptional and post-transcriptional regulation of LCN2 in health and disease. Furthermore, we summarize the impact of LCN2 dysregulation in a broad range of tumors. Lastly, we examine the mechanisms of action of LCN2 during tumorigenesis, progression, and metastasis. Understanding the complex relationships between LCN2 and tumor development, progression, and metastasis is vital for advancing our knowledge of cancer biology, developing biomarkers for diagnosis and clinical decision-making, and creating therapeutic strategies to improve the management of patients with cancer.
Collapse
Affiliation(s)
- BAOXING HUANG
- Clinical Laboratory, Shenzhen Children’s Hospital, Shenzhen, 518038, China
| | - ZICHANG JIA
- Clinical Laboratory, Shenzhen Children’s Hospital, Shenzhen, 518038, China
| | - CHENCHEN FU
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Key Laboratory of Tree Genetics and Biotechnology of Educational Department of China, Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Life Sciences, Nanjing Forestry University, Nanjing, 210037, China
| | - MOXIAN CHEN
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, Key Laboratory of Tree Genetics and Biotechnology of Educational Department of China, Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Life Sciences, Nanjing Forestry University, Nanjing, 210037, China
| | - ZEZHUO SU
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - YUNSHENG CHEN
- Clinical Laboratory, Shenzhen Children’s Hospital, Shenzhen, 518038, China
| |
Collapse
|
4
|
Paz AA, Jiménez TA, Ibarra-Gonzalez J, Astudillo-Maya C, Beñaldo FA, Figueroa EG, Llanos AJ, Gonzalez-Candia A, Herrera EA. Gestational hypoxia elicits long-term cardiovascular dysfunction in female guinea pigs. Life Sci 2025; 361:123282. [PMID: 39615619 DOI: 10.1016/j.lfs.2024.123282] [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: 09/05/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Gestational hypoxia (GH) has been implicated in the developmental programming of cardiovascular diseases (CVDs) in the offspring, with most studies focusing on males, conversely, the effects on female cardiovascular health remain understudied. We aimed to investigate the impact of GH on the cardiovascular system of female guinea pig offspring from the early postnatal period to adulthood. METHODS Pregnant guinea pigs were subjected to normoxic or hypoxic conditions from gestational day 30 until delivery (∼70 days). Female offspring were monitored with biometric parameters and peripheral vascular function (ultrasound) from birth to one year old. In addition, we assessed cardiovascular structure, oxidative stress, inflammatory state (IHC, qPCR, and immunoblot assays), and thoracic aorta reactivity (wire-myography) at one year of age. KEY FINDINGS GH increased heart rate and peripheral pulsatility index. At one year old, GH-exposed females exhibited cardiac remodeling, characterized by increased left ventricular luminal area and coronary artery muscle occupation. Furthermore, GH increased aortic vascular wall, intima-media thickness and contractile capacity. This was accompanied by reduced endothelium-dependent vasodilation and enhanced oxidative stress. Additionally, GH increased collagen deposition and oxidative stress in the right ventricle, accompanied by reduced antioxidant enzymes expression and reduced inflammatory mediator levels. SIGNIFICANCE GH exerts long-lasting effects on the cardiovascular health of female guinea pig offspring, contributing to cardiac remodeling, vascular dysfunction, oxidative stress, and inflammatory changes. These findings highlight the importance of GH as a risk factor for developing CVDs in female offspring and emphasize the need for sex-specific interventions to mitigate adverse long-term gestational effects.
Collapse
Affiliation(s)
- Adolfo A Paz
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Tamara A Jiménez
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Julieta Ibarra-Gonzalez
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Cristian Astudillo-Maya
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Felipe A Beñaldo
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Esteban G Figueroa
- Instituto de Ciencias de la Salud, Universidad de O'Higgins, Rancagua, Chile
| | - Aníbal J Llanos
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Putre, Chile
| | | | - Emilio A Herrera
- Laboratorio de Función y Reactividad Vascular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile; International Center for Andean Studies (INCAS), Universidad de Chile, Putre, Chile.
| |
Collapse
|
5
|
Alkaya M, Bayram A, Yaşar M, Doğan M, Gençer H. Lipocalin-2 expression in papillary thyroid carcinoma and its association with clinicopathological characteristics. Eur Arch Otorhinolaryngol 2025; 282:451-456. [PMID: 39242418 DOI: 10.1007/s00405-024-08954-w] [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: 02/26/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024]
Abstract
PURPOSE The present study aimed to assess Lipocalin-2 (LCN2) expression in patients with papillary thyroid cancer (PTC) and to compare it with multinodular goitre (MNG). We also investigated the correlation between LCN2 expression and clinicopathologic characteristics. METHODS This retrospective study included 63 surgically treated adult patients with papillary carcinoma and 65 adult patients with a MNG. Age, gender, physical, radiological and histopathological examinations, and surgical data of the patients were extracted from the hospital records. Size, histological subtype, capsule invasion, multifocality, extrathyroidal extension (ETE), lymph node metastasis (LNM), and immunohistochemical (IHC) studies of the tumour were recorded from the final histopathological reports of patients with PTC. The patient groups were compared in terms of LCN2 expression. The relationships between LCN2 expression and clinicopathological and other IHC parameters were also evaluated in patients with PTC. RESULTS LCN2 expression was significantly higher in the PTC group than in the control group. No significant correlation was demonstrated between LCN2 expression and the presence of multifocal disease, capsular invasion, vascular invasion, ETE, and LNM. There was a moderate positive correlation between LCN2 and human bone marrow endothelial cell marker-1 (HBME-1) expressions, however, no correlation was found between LCN2 and cytokeratin-19 (CK19), CD56, and galectin-3. CONCLUSION LCN2 expression may be a useful biomarker in differentiating benign and malignant lesions of the thyroid gland; however, its expression pattern may not be associated with clinicopathologic characteristics of the PTC and should be investigated in further studies with larger clinical samples.
Collapse
Affiliation(s)
- Mustafa Alkaya
- Department of ENT, Kayseri City Training and Research Hospital, Kayseri, Turkey.
- Halil Şıvgın State Hospital, Cumhuriyet Mh., Ankara Blv. No:54, 06760, Çubuk/Ankara, 05300, Turkey.
| | - Ali Bayram
- Department of ENT, Kayseri City Training and Research Hospital, Kayseri, Turkey
| | - Mehmet Yaşar
- Department of ENT, Kayseri City Training and Research Hospital, Kayseri, Turkey
| | - Merve Doğan
- Department of Pathology, Kayseri City Training and Research Hospital, Kayseri, Turkey
| | - Hümeyra Gençer
- Department of Nuclear Medicine, Kayseri City Training and Research Hospital, Kayseri, Turkey
| |
Collapse
|
6
|
Crescenzi E, Leonardi A, Pacifico F. NF-κB in Thyroid Cancer: An Update. Int J Mol Sci 2024; 25:11464. [PMID: 39519020 PMCID: PMC11546487 DOI: 10.3390/ijms252111464] [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: 09/23/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
The dysregulated NF-κB basal activity is a common feature of human thyroid carcinomas, especially in poorly differentiated or undifferentiated forms that, even if rare, are often resistant to standard therapies, and, therefore, are uncurable. Despite the molecular mechanisms leading to NF-κB activation in thyroid cancer being only partially understood, during the last few years, it has become clear that NF-κB contributes in different ways to the oncogenic potential of thyroid neoplastic cells. Indeed, it enhances their proliferation and viability, promotes their migration to and colonization of distant organs, and fuels their microenvironment. In addition, NF-κB signaling plays an important role in cancer stem cells from more aggressive thyroid carcinomas. Interfering with the different upstream and/or downstream pathways that drive NF-κB activity in thyroid neoplastic cells is an attractive strategy for the development of novel therapeutic drugs capable of overcoming the therapy resistance of advanced thyroid carcinomas. This review focuses on the recent findings about the key functions of NF-κB in thyroid cancer and discusses the potential implications of targeting NF-κB in advanced thyroid carcinomas.
Collapse
Affiliation(s)
- Elvira Crescenzi
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (CNR), Via S. Pansini, 5, 80131 Naples, Italy;
| | - Antonio Leonardi
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University of Naples Federico II, Via S. Pansini, 5, 80131 Naples, Italy;
| | - Francesco Pacifico
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (CNR), Via S. Pansini, 5, 80131 Naples, Italy;
| |
Collapse
|
7
|
Shi C, Wang C, Fu Z, Liu J, Zhou Y, Cheng B, Zhang C, Li S, Zhang Y. Lipocalin 2 (LCN2) confers acquired resistance to almonertinib in NSCLC through LCN2-MMP-9 signaling pathway. Pharmacol Res 2024; 201:107088. [PMID: 38295916 DOI: 10.1016/j.phrs.2024.107088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/05/2024]
Abstract
Almonertinib, a third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, is highly selective for EGFR-activating mutations as well as the EGFR T790M mutation in patients with advanced non-small cell lung cancer (NSCLC). However, the development of resistance inevitably occurs and poses a major obstacle to the clinical efficacy of almonertinib. Therefore, a clear understanding of the mechanism is of great significance to overcome drug resistance to almonertinib in the future. In this study, NCI-H1975 cell lines resistant to almonertinib (NCI-H1975 AR) were developed by concentration-increasing induction and were employed for clarification of underlying mechanisms of acquired resistance. Through RNA-seq analysis, the HIF-1 and TGF-β signaling pathways were significantly enriched by gene set enrichment analysis. Lipocalin-2 (LCN2), as the core node in these two signaling pathways, were found to be positively correlated to almonertinib-resistance in NSCLC cells. The function of LCN2 in the drug resistance of almonertinib was investigated through knockdown and overexpression assays in vitro and in vivo. Moreover, matrix metalloproteinases-9 (MMP-9) was further identified as a critical downstream effector of LCN2 signaling, which is regulated via the LCN2-MMP-9 axis. Pharmacological inhibition of MMP-9 could overcome resistance to almonertinib, as evidenced in both in vitro and in vivo models. Our findings suggest that LCN2 was a crucial regulator for conferring almonertinib-resistance in NSCLC and demonstrate the potential utility of targeting the LCN2-MMP-9 axis for clinical treatment of almonertinib-resistant lung adenocarcinoma.
Collapse
Affiliation(s)
- Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Cong Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinmei Liu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanfeng Zhou
- Department of Preclinical Translational Science, Shanghai Hansoh Biomedical Co.,Ltd., Shanghai 201203. China
| | - Bao Cheng
- Department of Chemistry, Shanghai Hansoh Biomedical Co., Ltd, Shanghai 201203, China
| | - Cong Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shijun Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China.
| |
Collapse
|
8
|
Crescenzi E, Mellone S, Gragnano G, Iaccarino A, Leonardi A, Pacifico F. NGAL Mediates Anaplastic Thyroid Carcinoma Cells Survival Through FAS/CD95 Inhibition. Endocrinology 2023; 165:bqad190. [PMID: 38091978 DOI: 10.1210/endocr/bqad190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Indexed: 12/27/2023]
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL), a siderophore-mediated iron binding protein, is highly expressed in human anaplastic thyroid carcinomas (ATCs) where it plays pleiotropic protumorigenic roles including that of a prosurvival protein. Here we show that NGAL inhibits FAS/CD95 death receptor to control ATC cell survival. FAS/CD95 expression in human specimens from patients with ATC and in ATC-derived cell lines negatively correlate with NGAL expression. Silencing of NGAL in ATC cells leads to FAS/CD95 upregulation, whereas NGAL overexpression determines the opposite effect. As a result, an agonist anti-FAS/CD95 antibody induces cell death in NGAL-silenced cells while it is ineffective on NGAL-overexpressing cells. Interestingly, the inhibitory activity of NGAL on FAS/CD95 is due to its iron carrier property given that perturbing iron homeostasis of NGAL-proficient and -deficient ATC cells directly influences FAS/CD95 expression. Accordingly, conditioned media containing a mutant form of NGAL unable to bind siderophores cannot rescue cells from FAS/CD95-dependent death, whereas NGAL wild type-containing conditioned media abolish the effects of the agonist antibody. We also find that downregulation of FAS/CD95 expression is mediated by iron-dependent NGAL suppression of p53 transcriptional activity. Our results indicate that NGAL contributes to ATC cell survival by iron-mediated inhibition of p53-dependent FAS/CD95 expression and suggest that restoring FAS/CD95 by NGAL suppression could be a helpful strategy to kill ATC cells.
Collapse
Affiliation(s)
- Elvira Crescenzi
- Istituto di Endocrinologia ed Oncologia Sperimentale, CNR, 80131 Naples, Italy
| | - Stefano Mellone
- Istituto di Endocrinologia ed Oncologia Sperimentale, CNR, 80131 Naples, Italy
| | - Gianluca Gragnano
- Dipartimento di Salute Pubblica, "Federico II" University of Naples, 80131 Naples, Italy
| | - Antonino Iaccarino
- Dipartimento di Salute Pubblica, "Federico II" University of Naples, 80131 Naples, Italy
| | - Antonio Leonardi
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, "Federico II" University of Naples, 80131 Naples, Italy
| | - Francesco Pacifico
- Istituto di Endocrinologia ed Oncologia Sperimentale, CNR, 80131 Naples, Italy
| |
Collapse
|
9
|
Cormier F, Housni S, Dumont F, Villard M, Cochand-Priollet B, Mercier-Nomé F, Perlemoine K, Bertherat J, Groussin L. NF-κB signaling activation and roles in thyroid cancers: implication of MAP3K14/NIK. Oncogenesis 2023; 12:55. [PMID: 37973791 PMCID: PMC10654696 DOI: 10.1038/s41389-023-00496-w] [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: 04/20/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023] Open
Abstract
Among follicular-derived thyroid cancers (TC), those with aggressive behavior and resistance to current treatments display poor prognosis. NF-κB signaling pathways are involved in tumor progression of various cancers. Here, we finely characterize the NF-κB pathways and their involvement in TC. By using immunoblot and gel shift assays, we demonstrated that both classical and alternative NF-κB pathways are activated in ten TC-derived cell lines, leading to activated RelA/p50 and RelB/p50 NF-κB dimers. By analyzing the RNAseq data of the large papillary thyroid carcinoma (PTC) cohort from The Cancer Genome Atlas (TCGA) project, we identified a tumor progression-related NF-κB signature in BRAFV600E mutated-PTCs. That corroborated with the role of RelA and RelB in cell migration and invasion processes that we demonstrated specifically in BRAFV600E mutated-cell lines, together with their role in the control of expression of genes implicated in invasiveness (MMP1, PLAU, LCN2 and LGALS3). We also identified NF-κB-inducing kinase (NIK) as a novel actor of the constitutive activation of the NF-κB pathways in TC-derived cell lines. Finally, its implication in invasiveness and its overexpression in PTC samples make NIK a potential therapeutic target for advanced TC treatment.
Collapse
Affiliation(s)
- Françoise Cormier
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France.
| | - Selma Housni
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France
- Service de Médecine Nucléaire, Assistance Publique-Hopitaux de Paris, Hopital Pitié-Salpêtrière, F-75013, Paris, France
| | - Florent Dumont
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France
- UMS IPSIT, Université Paris-Saclay, INSERM, CNRS, F-91400, Orsay, France
| | - Mélodie Villard
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France
| | - Béatrix Cochand-Priollet
- Service de Pathologie, Assistance Publique-Hopitaux de Paris, Hopital Cochin, Université Paris Cité, F-75014, Paris, France
| | | | - Karine Perlemoine
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France
| | - Jérôme Bertherat
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France
- Service d'Endocrinologie, Cochin AP-HP Centre, F-75014, Paris, France
| | - Lionel Groussin
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, F-75014, Paris, France
- Service d'Endocrinologie, Cochin AP-HP Centre, F-75014, Paris, France
| |
Collapse
|
10
|
Liu Y, Shao YH, Zhang JM, Wang Y, Zhou M, Li HQ, Zhang CC, Yu PJ, Gao SJ, Wang XR, Jia LX, Piao CM, Du J, Li YL. Macrophage CARD9 mediates cardiac injury following myocardial infarction through regulation of lipocalin 2 expression. Signal Transduct Target Ther 2023; 8:394. [PMID: 37828006 PMCID: PMC10570328 DOI: 10.1038/s41392-023-01635-w] [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: 02/16/2023] [Revised: 08/15/2023] [Accepted: 08/31/2023] [Indexed: 10/14/2023] Open
Abstract
Immune cell infiltration in response to myocyte death regulates extracellular matrix remodeling and scar formation after myocardial infarction (MI). Caspase-recruitment domain family member 9 (CARD9) acts as an adapter that mediates the transduction of pro-inflammatory signaling cascades in innate immunity; however, its role in cardiac injury and repair post-MI remains unclear. We found that Card9 was one of the most upregulated Card genes in the ischemic myocardium of mice. CARD9 expression increased considerably 1 day post-MI and declined by day 7 post-MI. Moreover, CARD9 was mainly expressed in F4/80-positive macrophages. Card9 knockout (KO) led to left ventricular function improvement and infarct scar size reduction in mice 28 days post-MI. Additionally, Card9 KO suppressed cardiomyocyte apoptosis in the border region and attenuated matrix metalloproteinase (MMP) expression. RNA sequencing revealed that Card9 KO significantly suppressed lipocalin 2 (Lcn2) expression post-MI. Both LCN2 and the receptor solute carrier family 22 member 17 (SL22A17) were detected in macrophages. Subsequently, we demonstrated that Card9 overexpression increased LCN2 expression, while Card9 KO inhibited necrotic cell-induced LCN2 upregulation in macrophages, likely through NF-κB. Lcn2 KO showed beneficial effects post-MI, and recombinant LCN2 diminished the protective effects of Card9 KO in vivo. Lcn2 KO reduced MMP9 post-MI, and Lcn2 overexpression increased Mmp9 expression in macrophages. Slc22a17 knockdown in macrophages reduced MMP9 release with recombinant LCN2 treatment. In conclusion, our results demonstrate that macrophage CARD9 mediates the deterioration of cardiac function and adverse remodeling post-MI via LCN2.
Collapse
Affiliation(s)
- Yan Liu
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yi-Hui Shao
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Jun-Meng Zhang
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Ying Wang
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Mei Zhou
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Hui-Qin Li
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Cong-Cong Zhang
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Pei-Jie Yu
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Shi-Juan Gao
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Xue-Rui Wang
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Li-Xin Jia
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Chun-Mei Piao
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yu-Lin Li
- Beijing Anzhen Hospital, Capital Medical University; The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing Collaborative Innovative Research Center for Cardiovascular Diseases; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China.
| |
Collapse
|
11
|
Crescenzi E, Leonardi A, Pacifico F. Iron Metabolism in Cancer and Senescence: A Cellular Perspective. BIOLOGY 2023; 12:989. [PMID: 37508419 PMCID: PMC10376531 DOI: 10.3390/biology12070989] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023]
Abstract
Iron participates in a number of biological processes and plays a crucial role in cellular homeostasis. Alterations in iron metabolism are considered hallmarks of cancer and drivers of aggressive behaviors, such as uncontrolled proliferation, resistance to apoptosis, enhanced metastatic ability, increased cell plasticity and stemness. Furthermore, a dysregulated iron metabolism has been associated with the development of an adverse tumor microenvironment. Alterations in iron metabolism have been described in cellular senescence and in aging. For instance, iron has been shown to accumulate in aged tissues and in age-related diseases. Furthermore, in vitro studies demonstrate increases in iron content in both replicative and stress-induced senescent cells. However, the role, the mechanisms of regulation and dysregulation and the effects of iron metabolism on senescence remain significantly less characterized. In this review, we first provide an overview of iron metabolism and iron regulatory proteins. Then, we summarize alterations in iron homeostasis in cancer and senescence from a cellular point of view.
Collapse
Affiliation(s)
- Elvira Crescenzi
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, CNR, Via S. Pansini, 5, 80131 Naples, Italy
| | - Antonio Leonardi
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, "Federico II" University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Francesco Pacifico
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, CNR, Via S. Pansini, 5, 80131 Naples, Italy
| |
Collapse
|
12
|
Romejko K, Markowska M, Niemczyk S. The Review of Current Knowledge on Neutrophil Gelatinase-Associated Lipocalin (NGAL). Int J Mol Sci 2023; 24:10470. [PMID: 37445650 DOI: 10.3390/ijms241310470] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL) is a 25-kDa protein that is secreted mostly by immune cells such as neutrophils, macrophages, and dendritic cells. Its production is stimulated in response to inflammation. The concentrations of NGAL can be measured in plasma, urine, and biological fluids such as peritoneal effluent. NGAL is known mainly as a biomarker of acute kidney injury and is released after tubular damage and during renal regeneration processes. NGAL is also elevated in chronic kidney disease and dialysis patients. It may play a role as a predictor of the progression of renal function decreases with complications and mortality due to kidney failure. NGAL is also useful in the diagnostic processes of cardiovascular diseases. It is highly expressed in injured heart tissue and atherosclerostic plaque; its serum concentrations correlate with the severity of heart failure and coronary artery disease. NGAL increases inflammatory states and its levels rise in arterial hypertension, obesity, diabetes, and metabolic complications such as insulin resistance, and is also involved in carcinogenesis. In this review, we present the current knowledge on NGAL and its involvement in different pathologies, especially its role in renal and cardiovascular diseases.
Collapse
Affiliation(s)
- Katarzyna Romejko
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine-National Research Institute, 128 Szaserów Street, 04-141 Warsaw, Poland
| | - Magdalena Markowska
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine-National Research Institute, 128 Szaserów Street, 04-141 Warsaw, Poland
| | - Stanisław Niemczyk
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine-National Research Institute, 128 Szaserów Street, 04-141 Warsaw, Poland
| |
Collapse
|
13
|
Barer L, Schröder SK, Weiskirchen R, Bacharach E, Ehrlich M. Lipocalin-2 regulates the expression of interferon-stimulated genes and the susceptibility of prostate cancer cells to oncolytic virus infection. Eur J Cell Biol 2023; 102:151328. [PMID: 37321037 DOI: 10.1016/j.ejcb.2023.151328] [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: 01/31/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/17/2023] Open
Abstract
Lipocalin-2 (LCN2) performs pleiotropic and tumor context-dependent functions in cancers of diverse etiologies. In prostate cancer (PCa) cells, LCN2 regulates distinct phenotypic features, including cytoskeleton organization and expression of inflammation mediators. Oncolytic virotherapy uses oncolytic viruses (OVs) to kill cancer cells and induce anti-tumor immunity. A main source of specificity of OVs towards tumor cells stems from cancer-induced defects in interferon (IFN)-based cell autonomous immune responses. However, the molecular underpinnings of such defects in PCa cells are only partially understood. Moreover, LCN2 effects on IFN responses of PCa cells and their susceptibility to OVs are unknown. To examine these issues, we queried gene expression databases for genes coexpressed with LCN2, revealing co-expression of IFN-stimulated genes (ISGs) and LCN2. Analysis of human PCa cells revealed correlated expression of LCN2 and subsets of IFNs and ISGs. CRISPR/Cas9-mediated stable knockout of LCN2 in PC3 cells or transient overexpression of LCN2 in LNCaP cells revealed LCN2-mediated regulation of IFNE (and IFNL1) expression, activation of JAK/STAT pathway, and expression of selected ISGs. Accordingly, and dependent on a functional JAK/STAT pathway, LCN2 reduced the susceptibility of PCa cells to infection with the IFN-sensitive OV, EHDV-TAU. In PC3 cells, LCN2 knockout increased phosphorylation of eukaryotic initiation factor 2α (p-eIF2α). Inhibition of PKR-like ER kinase (PERK) in PC3-LCN2-KO cells reduced p-eIF2α while increasing constitutive IFNE expression, phosphorylation of STAT1, and ISG expression; and decreasing EHDV-TAU infection. Together, these data propose that LCN2 regulates PCa susceptibility to OVs through attenuation of PERK activity and increased IFN and ISG expression.
Collapse
Affiliation(s)
- Lilach Barer
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Sarah K Schröder
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany.
| | - Eran Bacharach
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv-Yafo, Israel.
| | - Marcelo Ehrlich
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv-Yafo, Israel.
| |
Collapse
|
14
|
Di Paola R, De A, Capasso A, Giuliana S, Ranieri R, Ruosi C, Sciarra A, Vitagliano C, Perna AF, Capasso G, Simeoni M. Impact of Thyroid Cancer Treatment on Renal Function: A Relevant Issue to Be Addressed. J Pers Med 2023; 13:jpm13050813. [PMID: 37240983 DOI: 10.3390/jpm13050813] [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: 03/24/2023] [Revised: 05/02/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Thyroid cancers require complex and heterogeneous therapies with different impacts on renal function. In our systematic literature review, we analyzed several aspects: renal function assessment, the impact of radiotherapy and thyroid surgery on kidney functioning, and mechanisms of nephrotoxicity of different chemotherapy, targeted and immunologic drugs. Our study revealed that the renal impact of thyroid cancer therapy can be a limiting factor in all radiotherapy, surgery, and pharmacological approaches. It is advisable to conduct a careful nephrological follow-up imposing the application of body surface based estimated Glomerular Filtration Rate (eGFR) formulas for the purpose of an early diagnosis and treatment of renal failure, guaranteeing the therapy continuation to thyroid cancer patients.
Collapse
Affiliation(s)
- Rossella Di Paola
- Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Ananya De
- Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Anna Capasso
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas, Austin, TX 75063, USA
| | - Sofia Giuliana
- Nephrology Unit, Department of Specialist General Surgery, University Hospital "Luigi Vanvitelli", 80131 Naples, Italy
| | - Roberta Ranieri
- Nephrology Unit, Department of Specialist General Surgery, University Hospital "Luigi Vanvitelli", 80131 Naples, Italy
| | - Carolina Ruosi
- Nephrology Unit, Department of Specialist General Surgery, University Hospital "Luigi Vanvitelli", 80131 Naples, Italy
| | - Antonella Sciarra
- Department of Oncologic Surgery, Translational Medical Sciences at University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | - Caterina Vitagliano
- Nephrology Unit, Department of Specialist General Surgery, University Hospital "Luigi Vanvitelli", 80131 Naples, Italy
| | - Alessandra F Perna
- Nephrology and Dialysis Unit, Department of Translational Medical Sciences at University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | | | - Mariadelina Simeoni
- Nephrology and Dialysis Unit, Department of Translational Medical Sciences at University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| |
Collapse
|
15
|
Martiniakova M, Mondockova V, Biro R, Kovacova V, Babikova M, Zemanova N, Ciernikova S, Omelka R. The link between bone-derived factors osteocalcin, fibroblast growth factor 23, sclerostin, lipocalin 2 and tumor bone metastasis. Front Endocrinol (Lausanne) 2023; 14:1113547. [PMID: 36926025 PMCID: PMC10012867 DOI: 10.3389/fendo.2023.1113547] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/13/2023] [Indexed: 03/04/2023] Open
Abstract
The skeleton is the third most common site of metastatic disease, which causes serious bone complications and short-term prognosis in cancer patients. Prostate and breast cancers are responsible for the majority of bone metastasis, resulting in osteolytic or osteoblastic lesions. The crosstalk between bone cells and their interactions with tumor cells are important in the development of lesions. Recently, both preclinical and clinical studies documented the clinical relevance of bone-derived factors, including osteocalcin (OC) and its undercarboxylated form (ucOC), fibroblast growth factor 23 (FGF23), sclerostin (SCL), and lipocalin 2 (LCN2) as prognostic tumor biomarkers and potential therapeutic targets in bone metastasis. Both OC and ucOC could be useful targets for the prevention of bone metastasis in breast cancer. Moreover, elevated OC level may be a metastatic marker of prostate cancer. FGF23 is particularly important for those forms of cancer that primarily affect bone and/or are characterized by bone metastasis. In other tumor entities, increased FGF23 level is enigmatic. SCL plays a significant role in the pathogenesis of both osteolytic and osteoblastic lesions, as its levels are high in metastatic breast and prostate cancers. Elevated expression levels of LCN2 have been found in aggressive subtypes of cancer. However, its role in anti-metastasis varies significantly between different cancer types. Anyway, all aforementioned bone-derived factors can be used as promising tumor biomarkers. As metastatic bone disease is generally not curable, targeting bone factors represents a new trend in the prevention of bone metastasis and patient care.
Collapse
Affiliation(s)
- Monika Martiniakova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
- *Correspondence: Monika Martiniakova, ; Radoslav Omelka,
| | - Vladimira Mondockova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Roman Biro
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Veronika Kovacova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Martina Babikova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Nina Zemanova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, Bratislava, Slovakia
| | - Radoslav Omelka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Nitra, Slovakia
- *Correspondence: Monika Martiniakova, ; Radoslav Omelka,
| |
Collapse
|
16
|
Asaf S, Maqsood F, Jalil J, Sarfraz Z, Sarfraz A, Mustafa S, Ojeda IC. Lipocalin 2-not only a biomarker: a study of current literature and systematic findings of ongoing clinical trials. Immunol Res 2022; 71:287-313. [PMID: 36529828 PMCID: PMC9760530 DOI: 10.1007/s12026-022-09352-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Lipocalin 2 (Lcn2), also known as neutrophil gelatinase-associated lipocalin, is an innate immune protein encoded by the LCN2 gene. In this study, we investigated various roles and functions of Lcn2 characterized in a systems-based format and evaluated its therapeutic potentials and clinical relevance for diagnosis and prognosis. An additional systematic presentation was presented for 70 ongoing clinical trials utilizing Lcn2 in the diagnostic and prognostic setting as a key outcome measure. With trials being conducted through December 2030, Lcn2 will become all the more relevant given its associations with diseases as a prognostic biomarker. Data also suggests that it plays a role in pathological conditions. The gaps in our understanding of Lcn2, once filled, may improve the immune mediation of acute and chronic disease.
Collapse
Affiliation(s)
| | | | | | | | - Azza Sarfraz
- The Aga Khan University, Karachi, Pakistan.
- Department of Pediatrics and Child Health, Aga Khan University, Stadium Road, P.O Box 3500, Karachi, 74800, Pakistan.
| | | | | |
Collapse
|
17
|
Daniluk K, Lange A, Pruchniewski M, Małolepszy A, Sawosz E, Jaworski S. Delivery of Melittin as a Lytic Agent via Graphene Nanoparticles as Carriers to Breast Cancer Cells. J Funct Biomater 2022; 13:278. [PMID: 36547538 PMCID: PMC9787603 DOI: 10.3390/jfb13040278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/01/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Melittin, as an agent to lyse biological membranes, may be a promising therapeutic agent in the treatment of cancer. However, because of its nonspecific actions, there is a need to use a delivery method. The conducted research determined whether carbon nanoparticles, such as graphene and graphene oxide, could be carriers for melittin to breast cancer cells. The studies included the analysis of intracellular pH, the potential of cell membranes, the type of cellular transport, and the expression of receptor proteins. By measuring the particle size, zeta potential, and FT-IT analysis, we found that the investigated nanoparticles are connected by electrostatic interactions. The level of melittin encapsulation with graphene was 86%, while with graphene oxide it was 78%. A decrease in pHi was observed for all cell lines after administration of melittin and its complex with graphene. The decrease in membrane polarization was demonstrated for all lines treated with melittin and its complex with graphene and after exposure to the complex of melittin with graphene oxide for the MDA-MB-231 and HFFF2 lines. The results showed that the investigated melittin complexes and the melittin itself act differently on different cell lines (MDA-MB-231 and MCF-7). It has been shown that in MDA-MD-231 cells, melittin in a complex with graphene is transported to cells via caveolin-dependent endocytosis. On the other hand, the melittin-graphene oxide complex can reach breast cancer cells through various types of transport. Other differences in protein expression changes were also observed for tumor lines after exposure to melittin and complexes.
Collapse
Affiliation(s)
- Karolina Daniluk
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Agata Lange
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Michał Pruchniewski
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Artur Małolepszy
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, 00-654 Warsaw, Poland
| | - Ewa Sawosz
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Sławomir Jaworski
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| |
Collapse
|
18
|
Candido S, Tomasello B, Lavoro A, Falzone L, Gattuso G, Russo A, Paratore S, McCubrey JA, Libra M. Bioinformatic analysis of the LCN2-SLC22A17-MMP9 network in cancer: The role of DNA methylation in the modulation of tumor microenvironment. Front Cell Dev Biol 2022; 10:945586. [PMID: 36211450 PMCID: PMC9532607 DOI: 10.3389/fcell.2022.945586] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Several features of cancer cells such as proliferation, invasion, metastatic spreading, and drug resistance are affected by their interaction with several tumor microenvironment (TME) components, including neutrophil gelatinase-associated lipocalin (NGAL), solute carrier family 22 member 17 (SLC22A17), and matrix metallopeptidase 9 (MMP9). These molecules play a key role in tumor growth, invasion, and iron-dependent metabolism of cancer cells. However, the precise epigenetic mechanisms underlying the gene regulation of Lipocalin 2 (LCN2), SLC22A17, and MMP9 in cancer still remain unclear. To this purpose, computational analysis was performed on TCGA and GTEx datasets to evaluate the expression and DNA methylation status of LCN2, SLC22A17, and MMP9 genes in different tumor types. Correlation analysis between gene/isoforms expression and DNA methylation levels of LCN2, SLC22A17, and MMP9 was performed to investigate the role of DNA methylation in the modulation of these genes. Protein network analysis was carried out using reverse phase protein arrays (RPPA) data to identify protein-protein interactions of the LCN2-SLC22A17-MMP9 network. Furthermore, survival analysis was performed according to gene expression and DNA methylation levels. Our results demonstrated that LCN2 and MMP9 were mainly upregulated in most tumor types, whereas SLC22A17 was largely downregulated, representing a specific hallmark signature for all gastrointestinal tumors. Notably, the expression of LCN2, SLC22A17, and MMP9 genes was negatively affected by promoter methylation. Conversely, intragenic hypermethylation was associated with the overexpression of SLC22A17 and MMP9 genes. Protein network analysis highlighted the role of the LCN2-SLC22A17-MMP9 network in TME by the interaction with fibronectin 1 and claudin 7, especially in rectal tumors. Moreover, the impact of expression and methylation status of LCN2, SLC22A17, and MMP9 on overall survival and progression free interval was tumor type-dependent. Overall, our analyses provide a detailed overview of the expression and methylation status of LCN2, SLC22A17, and MMP9 in all TCGA tumors, indicating that the LCN2-SLC22A17-MMP9 network was strictly regulated by DNA methylation within TME. Our findings pave the way for the identification of novel DNA methylation hotspots with diagnostic and prognostic values and suitable for epi-drug targeting.
Collapse
Affiliation(s)
- Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, Catania, Italy
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Alessandro Lavoro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Luca Falzone
- Epidemiology Unit, IRCCS Istituto Nazionale Tumori “Fondazione G. Pascale”, Naples, Italy
| | - Giuseppe Gattuso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Angela Russo
- Pathological Anatomy Unit, ARNAS Garibaldi Hospital, Catania, Italy
| | - Sabrina Paratore
- Pathological Anatomy Unit, ARNAS Garibaldi Hospital, Catania, Italy
| | - James A. McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, Catania, Italy
| |
Collapse
|
19
|
Evaluation of Lipocalin-2 and Twist expression in thyroid cancers and its relationship with epithelial mesenchymal transition. Ann Diagn Pathol 2022; 59:151973. [DOI: 10.1016/j.anndiagpath.2022.151973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/16/2022] [Accepted: 05/05/2022] [Indexed: 11/19/2022]
|
20
|
Joseph DB, Henry GH, Malewska A, Reese JC, Mauck RJ, Gahan JC, Hutchinson RC, Mohler JL, Roehrborn CG, Strand DW. 5-alpha reductase inhibitors induce a prostate luminal to club cell transition in human benign prostatic hyperplasia. J Pathol 2021; 256:427-441. [PMID: 34928497 DOI: 10.1002/path.5857] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/22/2021] [Accepted: 12/17/2021] [Indexed: 11/09/2022]
Abstract
Benign prostatic hyperplasia (BPH) is a progressive expansion of peri-urethral prostate tissue common in aging men. Patients with enlarged prostates are treated with 5-alpha reductase inhibitors (5ARIs) to shrink prostate volume by blocking the conversion of testosterone to dihydrotestosterone (DHT). A reduction in DHT levels can elicit atrophy and apoptosis of prostate secretory luminal cells, which results in a favorable clinical response characterized by improved lower urinary tract symptoms. However, the histologic response to 5ARI treatment is often heterogeneous across prostate acini and lower urinary tract symptoms can persist to require surgical intervention. We used two spatial profiling approaches to characterize gene expression changes across histologically normal and atrophied regions in prostates from 5ARI-treated men. Objective transcriptomic profiling using the Visium spatial gene expression platform showed that 5ARI-induced atrophy of prostate luminal cells correlated with reduced androgen receptor signaling and increased expression of urethral club cell genes including LTF, PIGR, OLFM4, SCGB1A1 and SCGB3A1. Prostate luminal cells within atrophied acini adapted to decreased DHT conditions by increasing NF-κB signaling and anti-apoptotic BCL2 expression, which may explain their survival. Using GeoMx digital spatial profiling with a probe set to assess ~18,000 RNA targets, we confirmed that atrophied acini expressing SCGB3A1 displayed higher levels of club cell markers compared to histologically normal acini with NKX3-1 expression. In addition, club-like cells within regions of 5ARI-induced atrophy closely resembled true club cells from the prostatic urethra. A comparison of histologically normal regions from 5ARI-treated men and histologically normal regions from untreated men revealed few transcriptional differences. Taken together, our results describe a heterogeneous response to 5ARI treatment where cells in atrophied acini undergo an adaptation from a prostate secretory luminal to a club cell-like state in response to 5ARI treatment. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Diya B Joseph
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Gervaise H Henry
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Alicia Malewska
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Ryan J Mauck
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey C Gahan
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ryan C Hutchinson
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - James L Mohler
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Claus G Roehrborn
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Douglas W Strand
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
21
|
Crosstalk between E-Cadherin/β-Catenin and NF-κB Signaling Pathways: The Regulation of Host-Pathogen Interaction during Leptospirosis. Int J Mol Sci 2021; 22:ijms222313132. [PMID: 34884937 PMCID: PMC8658460 DOI: 10.3390/ijms222313132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/29/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023] Open
Abstract
Approximately 1 million cases of leptospirosis, an emerging infectious zoonotic disease, are reported each year. Pathogenic Leptospira species express leucine-rich repeat (LRR) proteins that are rarely expressed in non-pathogenic Leptospira species. The LRR domain-containing protein family is vital for the virulence of pathogenic Leptospira species. In this study, the biological mechanisms of an essential LRR domain protein from pathogenic Leptospira were examined. The effects of Leptospira and recombinant LRR20 (rLRR20) on the expression levels of factors involved in signal transduction were examined using microarray, quantitative real-time polymerase chain reaction, and western blotting. The secreted biomarkers were measured using an enzyme-linked immunosorbent assay. rLRR20 colocalized with E-cadherin on the cell surface and activated the downstream transcription factor β-catenin, which subsequently promoted the expression of MMP7, a kidney injury biomarker. Additionally, MMP7 inhibitors were used to demonstrate that the secreted MMP7 degrades surface E-cadherin. This feedback inhibition mechanism downregulated surface E-cadherin expression and inhibited the colonization of Leptospira. The degradation of surface E-cadherin activated the NF-κB signal transduction pathway. Leptospirosis-associated acute kidney injury is associated with the secretion of NGAL, a downstream upregulated biomarker of the NF-κB signal transduction pathway. A working model was proposed to illustrate the crosstalk between E-cadherin/β-catenin and NF-κB signal transduction pathways during Leptospira infection. Thus, rLRR20 of Leptospira induces kidney injury in host cells and inhibits the adhesion and invasion of Leptospira through the upregulation of MMP7 and NGAL.
Collapse
|
22
|
NGAL as a Potential Target in Tumor Microenvironment. Int J Mol Sci 2021. [DOI: 10.3390/ijms222212333
expr 804735418 + 979474750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
The signaling network between cancer and stromal cells plays a crucial role in tumor microenvironment. The fate of tumor progression mainly depends on the huge amount of information that these cell populations exchange from the onset of neoplastic transformation. Interfering with such signaling has been producing exciting results in cancer therapy: just think of anti-PD-1/anti-PD-L1/anti-CTLA-4 antibodies that, acting as immune checkpoint inhibitors, interrupt the inhibitory signaling exerted by cancer cells on immune cells or the CAR-T technology that fosters the reactivation of anti-tumoral immunity in a restricted group of leukemias and lymphomas. Nevertheless, many types of cancers, in particular solid tumors, are still refractory to these treatments, so the identification of novel molecular targets in tumor secretome would benefit from implementation of current anti-cancer therapeutical strategies. Neutrophil Gelatinase-Associated Lipocalin (NGAL) is a secreted protein abundantly expressed in the secretome of various human tumors. It represents a promising target for the multiple roles that are played inside cancer and stromal cells, and also overall in their cross-talk. The review focuses on the different roles of NGAL in tumor microenvironment and in cancer senescence-associated secretory phenotype (SASP), highlighting the most crucial functions that could be eventually targetable in cancer therapy.
Collapse
|
23
|
Crescenzi E, Leonardi A, Pacifico F. NGAL as a Potential Target in Tumor Microenvironment. Int J Mol Sci 2021; 22:12333. [PMID: 34830212 PMCID: PMC8623964 DOI: 10.3390/ijms222212333&set/a 915137580+984946846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
The signaling network between cancer and stromal cells plays a crucial role in tumor microenvironment. The fate of tumor progression mainly depends on the huge amount of information that these cell populations exchange from the onset of neoplastic transformation. Interfering with such signaling has been producing exciting results in cancer therapy: just think of anti-PD-1/anti-PD-L1/anti-CTLA-4 antibodies that, acting as immune checkpoint inhibitors, interrupt the inhibitory signaling exerted by cancer cells on immune cells or the CAR-T technology that fosters the reactivation of anti-tumoral immunity in a restricted group of leukemias and lymphomas. Nevertheless, many types of cancers, in particular solid tumors, are still refractory to these treatments, so the identification of novel molecular targets in tumor secretome would benefit from implementation of current anti-cancer therapeutical strategies. Neutrophil Gelatinase-Associated Lipocalin (NGAL) is a secreted protein abundantly expressed in the secretome of various human tumors. It represents a promising target for the multiple roles that are played inside cancer and stromal cells, and also overall in their cross-talk. The review focuses on the different roles of NGAL in tumor microenvironment and in cancer senescence-associated secretory phenotype (SASP), highlighting the most crucial functions that could be eventually targetable in cancer therapy.
Collapse
Affiliation(s)
- Elvira Crescenzi
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale, CNR, Via S. Pansini, 5-80131 Naples, Italy;
| | - Antonio Leonardi
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, “Federico II” University of Naples, Via S. Pansini, 5-80131 Naples, Italy;
| | - Francesco Pacifico
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale, CNR, Via S. Pansini, 5-80131 Naples, Italy;
- Correspondence:
| |
Collapse
|
24
|
NGAL as a Potential Target in Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms222212333. [PMID: 34830212 PMCID: PMC8623964 DOI: 10.3390/ijms222212333] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 12/29/2022] Open
Abstract
The signaling network between cancer and stromal cells plays a crucial role in tumor microenvironment. The fate of tumor progression mainly depends on the huge amount of information that these cell populations exchange from the onset of neoplastic transformation. Interfering with such signaling has been producing exciting results in cancer therapy: just think of anti-PD-1/anti-PD-L1/anti-CTLA-4 antibodies that, acting as immune checkpoint inhibitors, interrupt the inhibitory signaling exerted by cancer cells on immune cells or the CAR-T technology that fosters the reactivation of anti-tumoral immunity in a restricted group of leukemias and lymphomas. Nevertheless, many types of cancers, in particular solid tumors, are still refractory to these treatments, so the identification of novel molecular targets in tumor secretome would benefit from implementation of current anti-cancer therapeutical strategies. Neutrophil Gelatinase-Associated Lipocalin (NGAL) is a secreted protein abundantly expressed in the secretome of various human tumors. It represents a promising target for the multiple roles that are played inside cancer and stromal cells, and also overall in their cross-talk. The review focuses on the different roles of NGAL in tumor microenvironment and in cancer senescence-associated secretory phenotype (SASP), highlighting the most crucial functions that could be eventually targetable in cancer therapy.
Collapse
|
25
|
Wang F, Zhang C, Cheng H, Liu C, Lu Z, Zheng S, Wang S, Sun N, He J. TGF-β-induced PLEK2 promotes metastasis and chemoresistance in oesophageal squamous cell carcinoma by regulating LCN2. Cell Death Dis 2021; 12:901. [PMID: 34601488 PMCID: PMC8487427 DOI: 10.1038/s41419-021-04155-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
Oesophageal squamous cell carcinoma (ESCC) has a relatively unfavourable prognosis due to metastasis and chemoresistance. Our previous research established a comprehensive ESCC database (GSE53625). After analysing data from TCGA database and GSE53625, we found that PLEK2 predicted poor prognosis in ESCC. Moreover, PLEK2 expression was also related to the overall survival of ESCC patients undergoing chemotherapy. Repression of PLEK2 decreased the proliferation, migration, invasion and chemoresistance of ESCC cells in vitro and decreased tumorigenicity and distant metastasis in vivo. Mechanistically, luciferase reporter assay and chromatin immunoprecipitation assay suggested that TGF-β stimulated the process that Smad2/3 binds to the promoter sequences of PLEK2 and induced its expression. RNA-seq suggested LCN2 might a key molecular regulated by PLEK2. LCN2 overexpression in PLEK2 knockdown ESCC cells reversed the effects of decreased migration and invasion. In addition, TGF-β induced the expression of LCN2, but the effect disappeared when PLEK2 was knockdown. Moreover, AKT was phosphorylated in all regulatory processes. This study detected the major role of PLEK2 in driving metastasis and chemoresistance in ESCC by regulating LCN2, which indicates the potential use of PLEK2 as a biomarker to predict prognosis and as a therapeutic target for ESCC.
Collapse
Affiliation(s)
- Feng Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong Cheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chengming Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiliang Lu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sufei Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sihui Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing,, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
26
|
Villodre ES, Hu X, Larson R, Finetti P, Gomez K, Balema W, Stecklein SR, Santiago‐Sanchez G, Krishnamurthy S, Song J, Su X, Ueno NT, Tripathy D, Van Laere S, Bertucci F, Vivas‐Mejía P, Woodward WA, Debeb BG. Lipocalin 2 promotes inflammatory breast cancer tumorigenesis and skin invasion. Mol Oncol 2021; 15:2752-2765. [PMID: 34342930 PMCID: PMC8486564 DOI: 10.1002/1878-0261.13074] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/21/2021] [Accepted: 08/02/2021] [Indexed: 12/28/2022] Open
Abstract
Inflammatory breast cancer (IBC) is an aggressive form of primary breast cancer characterized by rapid onset and high risk of metastasis and poor clinical outcomes. The biological basis for the aggressiveness of IBC is still not well understood and no IBC-specific targeted therapies exist. In this study, we report that lipocalin 2 (LCN2), a small secreted glycoprotein belonging to the lipocalin superfamily, is expressed at significantly higher levels in IBC vs non-IBC tumors, independently of molecular subtype. LCN2 levels were also significantly higher in IBC cell lines and in their culture media than in non-IBC cell lines. High expression was associated with poor-prognosis features and shorter overall survival in IBC patients. Depletion of LCN2 in IBC cell lines reduced colony formation, migration, and cancer stem cell populations in vitro and inhibited tumor growth, skin invasion, and brain metastasis in mouse models of IBC. Analysis of our proteomics data showed reduced expression of proteins involved in cell cycle and DNA repair in LCN2-silenced IBC cells. Our findings support that LCN2 promotes IBC tumor aggressiveness and offer a new potential therapeutic target for IBC.
Collapse
Affiliation(s)
- Emilly S. Villodre
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Xiaoding Hu
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Richard Larson
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Pascal Finetti
- Laboratory of Predictive OncologyAix‐Marseille UniversityInsermCNRSInstitut Paoli‐CalmettesCRCMMarseilleFrance
| | - Kristen Gomez
- Department of Biological SciencesThe University of Texas at BrownsvilleTXUSA
| | - Wintana Balema
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Shane R. Stecklein
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Ginette Santiago‐Sanchez
- Department Biochemistry and Cancer CenterUniversity of Puerto Rico Medical Sciences CampusSan Juan, Puerto Rico
| | - Savitri Krishnamurthy
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of PathologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Juhee Song
- Department of BiostatisticsThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Xiaoping Su
- Department of Bioinformatics and Computational BiologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Naoto T. Ueno
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Debu Tripathy
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Steven Van Laere
- Center for Oncological Research (CORE)Integrated Personalized and Precision Oncology Network (IPPON)University of AntwerpBelgium
| | - François Bertucci
- Laboratory of Predictive OncologyAix‐Marseille UniversityInsermCNRSInstitut Paoli‐CalmettesCRCMMarseilleFrance
| | - Pablo Vivas‐Mejía
- Department Biochemistry and Cancer CenterUniversity of Puerto Rico Medical Sciences CampusSan Juan, Puerto Rico
| | - Wendy A. Woodward
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Bisrat G. Debeb
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| |
Collapse
|
27
|
Umar MI, Hassan W, Murtaza G, Buabeid M, Arafa E, Irfan HM, Asmawi MZ, Huang X. The Adipokine Component in the Molecular Regulation of Cancer Cell Survival, Proliferation and Metastasis. Pathol Oncol Res 2021; 27:1609828. [PMID: 34588926 PMCID: PMC8473628 DOI: 10.3389/pore.2021.1609828] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/09/2021] [Indexed: 12/22/2022]
Abstract
A hormonal imbalance may disrupt the rigorously monitored cellular microenvironment by hampering the natural homeostatic mechanisms. The most common example of such hormonal glitch could be seen in obesity where the uprise in adipokine levels is in virtue of the expanding bulk of adipose tissue. Such aberrant endocrine signaling disrupts the regulation of cellular fate, rendering the cells to live in a tumor supportive microenvironment. Previously, it was believed that the adipokines support cancer proliferation and metastasis with no direct involvement in neoplastic transformations and tumorigenesis. However, the recent studies have reported discrete mechanisms that establish the direct involvement of adipokine signaling in tumorigenesis. Moreover, the individual adipokine profile of the patients has never been considered in the prognosis and staging of the disease. Hence, the present manuscript has focused on the reported extensive mechanisms that culminate the basis of poor prognosis and diminished survival rate in obese cancer patients.
Collapse
Affiliation(s)
| | - Waseem Hassan
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Manal Buabeid
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Elshaimaa Arafa
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | | | - Mohd Zaini Asmawi
- School of Pharmaceutical Sciences, University of Science Malaysia, Pulau Pinang, Malaysia
| | - Xianju Huang
- College of Pharmacy, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
28
|
Pourmohammadi-Bejarpasi Z, Sabzevari R, Mohammadi Roushandeh A, Ebrahimi A, Mobayen M, Jahanian-Najafabadi A, Darjani A, Habibi Roudkenar M. Combination Therapy of Metadichol Nanogel and Lipocalin-2 Engineered Mesenchymal Stem Cells Improve Wound Healing in Rat Model of Excision Injury. Adv Pharm Bull 2021; 12:550-560. [PMID: 35935055 PMCID: PMC9348536 DOI: 10.34172/apb.2022.059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/29/2021] [Accepted: 09/11/2021] [Indexed: 11/11/2022] Open
Abstract
Purpose: Currently, several disorders including burns, trauma, excisional and diabetic wounds, and bedsores threaten the human health. Application of mesenchymal stem cells (MSCs) is recommended for treatment of skin disorders. However, because of oxidative stress and inflammation after skin injury, survival of transplanted MSCs is low which in turn negatively affects the efficiency of the MSCs-based therapy. In an attempt to address the aforementioned challenge and introducing a novel potential therapeutic strategy, we employed combination therapy by lipocalin 2 (Lcn2)-engineered MSCs and a Metadichol (an inverse agonist of vitamin D receptor (VDR)) nanogel in a rat model of excisional wound.
Methods: First, human umbilical cord MSCs (hUC-MSCs) was transfected by a recombinant plasmid encoding Lcn2 gene. Next, a combination of Metadichol nanogel and the engineered MSCs was co-applied on wound in rat model of excision injury. Finally the improvement of wound healing in experimental groups was evaluated by photography and histological assessments (hematoxylin and eosin staining).
Results: Our findings revealed that the repair rate was higher in the group received combination therapy comparing to control groups. Notably, Metadichol+Lcn2-MSCs showed significantly higher wound contraction rate compared to control group at all time points (P value < 0.001). Furthermore, wound repair rate was 95% 14 days after surgery, and 100% after 21 days in the treatment groups. Our results also revealed that the combination therapy improved and accelerated the wound healing process.
Conclusion: Our findings suggest a novel potential therapeutic strategy i.e. Lcn2-engineered MSCs and Metadichol for wound healing. However, further preclinical and clinical studies are required.
Collapse
Affiliation(s)
| | - Reza Sabzevari
- Medical Biotechnology Department, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| | - Amaneh Mohammadi Roushandeh
- Cellular and Molecular Research Center, Medicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
- Burn and Regenerative Research Center, Medicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| | - Ammar Ebrahimi
- Medical Biotechnology Department, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammadreza Mobayen
- Burn and Regenerative Research Center, Medicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Jahanian-Najafabadi
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Abbas Darjani
- Skin Research Center, Department of Dermatology, Razi Hospital, School of Medicine, Guilan University of Medical Science, Rasht, Iran
| | - Mehryar Habibi Roudkenar
- Cellular and Molecular Research Center, Medicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
- Burn and Regenerative Research Center, Medicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
29
|
Tonnus W, Belavgeni A, Beuschlein F, Eisenhofer G, Fassnacht M, Kroiss M, Krone NP, Reincke M, Bornstein SR, Linkermann A. The role of regulated necrosis in endocrine diseases. Nat Rev Endocrinol 2021; 17:497-510. [PMID: 34135504 PMCID: PMC8207819 DOI: 10.1038/s41574-021-00499-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 12/13/2022]
Abstract
The death of endocrine cells is involved in type 1 diabetes mellitus, autoimmunity, adrenopause and hypogonadotropism. Insights from research on basic cell death have revealed that most pathophysiologically important cell death is necrotic in nature, whereas regular metabolism is maintained by apoptosis programmes. Necrosis is defined as cell death by plasma membrane rupture, which allows the release of damage-associated molecular patterns that trigger an immune response referred to as necroinflammation. Regulated necrosis comes in different forms, such as necroptosis, pyroptosis and ferroptosis. In this Perspective, with a focus on the endocrine environment, we introduce these cell death pathways and discuss the specific consequences of regulated necrosis. Given that clinical trials of necrostatins for the treatment of autoimmune conditions have already been initiated, we highlight the therapeutic potential of such novel therapeutic approaches that, in our opinion, should be tested in endocrine disorders in the future.
Collapse
Affiliation(s)
- Wulf Tonnus
- Clinic of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Alexia Belavgeni
- Clinic of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV, Hospital of the Ludwig-Maximilian-University Munich, Munich, Germany
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zürich, Switzerland
| | - Graeme Eisenhofer
- Clinic of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Martin Fassnacht
- Clinic of Internal Medicine I, Division of Endocrinology and Diabetology, University Hospital, University of Würzburg, Würzburg, Germany
| | - Matthias Kroiss
- Clinic of Internal Medicine I, Division of Endocrinology and Diabetology, University Hospital, University of Würzburg, Würzburg, Germany
| | - Nils P Krone
- Clinic of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Academic Unit of Child Health, Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Hospital of the Ludwig-Maximilian-University Munich, Munich, Germany
| | - Stefan R Bornstein
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zürich, Switzerland
- Clinic of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Diabetes and Nutritional Sciences, King's College London, London, UK
| | - Andreas Linkermann
- Clinic of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
30
|
Lu KH, Yang JS, Hsieh YH, Chu HJ, Chou CH, Lu EWH, Lin CW, Yang SF. Lipocalin-2 Inhibits Osteosarcoma Cell Metastasis by Suppressing MET Expression via the MEK-ERK Pathway. Cancers (Basel) 2021; 13:cancers13133181. [PMID: 34202288 PMCID: PMC8268143 DOI: 10.3390/cancers13133181] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Higher neutrophil-derived cytokine lipocalin-2 (LCN2) expression possesses a versatile role in a myriad of cancers, but little is known about the role of LCN2 on osteosarcoma metastasis. In this study, we demonstrated that higher LCN2 inhibited cellular motility, migration, and invasion of osteosarcoma cells. Moreover, the phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 was decreased by LCN2 knockdown. Conclusively, LCN2 inhibits osteosarcoma cell metastasis by suppressing MET via the mitogen-activated protein kinases/ERK kinase (MEK)–ERK pathway. Abstract Higher neutrophil-derived cytokine lipocalin-2 (LCN2) expression possesses a versatile role in a myriad of cancers, but little is known about the role of LCN2 on osteosarcoma metastasis. In this study, we demonstrated that higher LCN2 inhibited cellular motility, migration, and invasion of osteosarcoma cells. Moreover, using RNA sequencing technology, we found that LCN2 repressed MET gene expression in U2OS cells. Manipulation of LCN2 levels influenced the migratory potential of osteosarcoma cells as cellular migration was enhanced by transfecting with vectors containing a constitutively active LCN2 cDNA and recombinant human LCN2. Moreover, the phosphorylation of mitogen-activated protein kinases/extracellular signal-regulated kinase (ERK) kinase (MEK) 1/2 and ERK 1/2 was decreased by LCN2 knockdown. Furthermore, the use of ERK inhibitor (U0126) and activator (tBHQ) confirmed that the pharmaceutic inhibition of MEK–ERK augmented the LCN2-mediated MET suppression and migration of U2OS and HOS cells. Conclusively, LCN2 inhibits osteosarcoma cell metastasis by suppressing MET via the MEK–ERK pathway.
Collapse
Affiliation(s)
- Ko-Hsiu Lu
- Department of Orthopedics, Chung Shan Medical University Hospital, Taichung 402, Taiwan;
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Jia-Sin Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (J.-S.Y.); (Y.-H.H.); (H.-J.C.); (C.-H.C.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (J.-S.Y.); (Y.-H.H.); (H.-J.C.); (C.-H.C.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Hsiao-Ju Chu
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (J.-S.Y.); (Y.-H.H.); (H.-J.C.); (C.-H.C.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Chia-Hsuan Chou
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (J.-S.Y.); (Y.-H.H.); (H.-J.C.); (C.-H.C.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | | | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- Correspondence: (C.-W.L.); (S.-F.Y.); Tel.: +886-4-24739595-34253 (S.-F.Y)
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (J.-S.Y.); (Y.-H.H.); (H.-J.C.); (C.-H.C.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- Correspondence: (C.-W.L.); (S.-F.Y.); Tel.: +886-4-24739595-34253 (S.-F.Y)
| |
Collapse
|
31
|
Liang W, Ferrara N. Iron Metabolism in the Tumor Microenvironment: Contributions of Innate Immune Cells. Front Immunol 2021; 11:626812. [PMID: 33679721 PMCID: PMC7928394 DOI: 10.3389/fimmu.2020.626812] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/30/2020] [Indexed: 12/21/2022] Open
Abstract
Cells of the innate immune system are a major component of the tumor microenvironment. They play complex and multifaceted roles in the regulation of cancer initiation, growth, metastasis and responses to therapeutics. Innate immune cells like neutrophils and macrophages are recruited to cancerous tissues by chemotactic molecules released by cancer cells and cancer-associated stromal cells. Once they reach the tumor, they can be instructed by a network of proteins, nucleic acids and metabolites to exert protumoral or antitumoral functions. Altered iron metabolism is a feature of cancer. Epidemiological studies suggest that increased presence of iron and/or iron binding proteins is associated with increased risks of cancer development. It has been shown that iron metabolism is involved in shaping the immune landscapes in inflammatory/infectious diseases and cancer-associated inflammation. In this article, we will dissect the contribution of macrophages and neutrophils to dysregulated iron metabolism in malignant cells and its impact on cancer growth and metastasis. The mechanisms involved in regulating the actions of macrophages and neutrophils will also be discussed. Moreover, we will examine the effects of iron metabolism on the phenotypes of innate immune cells. Both iron chelating and overloading agents are being explored in cancer treatment. This review highlights alternative strategies for management of iron content in cancer cells by targeting the iron donation and modulation properties of macrophages and neutrophils in the tumor microenvironment.
Collapse
Affiliation(s)
- Wei Liang
- Oncology, BioDuro LLC, San Diego, CA, United States
| | - Napoleone Ferrara
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
32
|
Zhang MX, Wang L, Zeng L, Tu ZW. LCN2 Is a Potential Biomarker for Radioresistance and Recurrence in Nasopharyngeal Carcinoma. Front Oncol 2021; 10:605777. [PMID: 33604288 PMCID: PMC7885862 DOI: 10.3389/fonc.2020.605777] [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: 09/13/2020] [Accepted: 12/15/2020] [Indexed: 12/24/2022] Open
Abstract
Background Radioresistance-induced local failure, which can result in residual or recurrent tumors, remains one of the major causes of treatment failure in nasopharyngeal carcinoma (NPC). Lipocalin 2 (LCN2) is known to play important roles in cancer initiation, progression, and treatment responses. However, its role in the radioresistance of NPC remains unclear. Methods Microarray data from the Gene Expression Omnibus (GEO) was screened for candidate biomarkers relating to the radioresistance of NPC. The expression of LCN2 in NPC cell lines was verified by quantitative real-time PCR (RT-qPCR) and western blotting. The effects of knockdown or overexpression of LCN2 on NPC radiosensitivity were examined using a soft agar colony formation assay and a γH2AX assay. LCN2 expression in NPC specimens was evaluated by immunohistochemistry. Survival outcomes were analyzed. A possible correlation between LCN2 and hypoxia-inducible factor 1-alpha (HIF-1A) was examined by western blotting and a tissue microarray. Results LCN2 was highly expressed in the radioresistant NPC cell line CNE2R. Knocking down LCN2 enhanced the radiosensitivity of NPC cells by impairing their ability to repair DNA damage or proliferate, while ectopic expression of LCN2 conferred additional radioresistance to NPC cells. Immunohistochemical analysis of 100 NPC specimens revealed that LCN2 expression was significantly upregulated in radioresistant NPC tissues and was associated with NPC recurrence. Furthermore, a significant correlation between the expression of LCN2 and HIF-1A was detected. Conclusion LCN2 is associated with radioresistance and recurrence in NPC and may facilitate the development of a radioresistant phenotype through interacting with HIF-1A. Our data indicate that LCN2 is a promising target for predicting and overcoming radioresistance in NPC.
Collapse
Affiliation(s)
- Meng-Xia Zhang
- State Key Laboratory of Oncology in South China, Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Li Wang
- Department of Radiotherapy, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Lei Zeng
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zi-Wei Tu
- NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma (Jiangxi Cancer Hospital of Nanchang University), Nanchang, China
| |
Collapse
|
33
|
Che K, Han W, Zhang M, Niu H. Role of neutrophil gelatinase-associated lipocalin in renal cell carcinoma. Oncol Lett 2021; 21:148. [PMID: 33552266 PMCID: PMC7798090 DOI: 10.3892/ol.2020.12409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/26/2020] [Indexed: 01/03/2023] Open
Abstract
Human neutrophil gelatinase-associated lipocalin (NGAL) is a glycoprotein present in a wide variety of tissues and cell types. It exists as a monomer of 25 kDa, a homodimer of 45 kDa or a heterodimer of 135 kDa (disulfide bound to latent matrix metalloproteinase-9). NGAL is considered the biochemical gold standard for the early diagnosis of acute kidney injury and has attracted much attention as a diagnostic biomarker. NGAL has controversial (i.e. both beneficial and detrimental) effects on cellular processes associated with tumor development, such as cell proliferation, survival, migration, invasion and drug resistance. Therefore, the present review aimed at clarifying the role of NGAL in renal cell carcinoma (RCC). Relevant studies of NGAL and RCC were searched in PubMed and relevant information about the structure, expression, function and mechanism of NGAL in RCC were summarized. Finally, the following conclusions could be drawn from the literature: i) NGAL can be detected in cancer tissues, serum and urine of patients with RCC; ii) NGAL is not a suitable diagnostic marker for early screening of RCC; iii) NGAL expression may be used to predict the prognosis of patients with RCC; and iv) Further research on NGAL may be helpful to decrease sunitinib resistance and find new treatment strategies for RCC.
Collapse
Affiliation(s)
- Kai Che
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
- Department of Clinical Medicine, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Wenkai Han
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
- Department of Clinical Medicine, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Mingxin Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Haitao Niu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
34
|
Metovic J, Vignale C, Annaratone L, Osella-Abate S, Maletta F, Rapa I, Cabutti F, Patriarca S, Gallo M, Nikiforov YE, Volante M, Papotti M. The Oncocytic Variant of Poorly Differentiated Thyroid Carcinoma Shows a Specific Immune-Related Gene Expression Profile. J Clin Endocrinol Metab 2020; 105:5906602. [PMID: 32936917 DOI: 10.1210/clinem/dgaa655] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 09/14/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Poorly differentiated thyroid cancer (PDTC) is a rare, follicular cell-derived neoplasm with an unfavorable prognosis. The oncocytic variant of PDTC may be associated with even more adverse outcome than classical PDTC cases, but its specific molecular features are largely unknown. Our aim was to explore the immune-related gene expression profile of oncocytic and classical PDTC, in correlation with clinical and pathological characteristics (including programmed death ligand 1 [PD-L1] expression) and outcome, and in comparison with a control group of well-differentiated follicular carcinomas (WDFCs), including conventional follicular carcinomas (FTCs) and Hürthle cell carcinomas (HCCs). METHODS A retrospective series of 48 PDTCs and 24 WDFCs was analyzed by means of NanoString technology employing the nCounter PanCancer Immune Profiling panel. Gene expression data were validated using quantitative real-time polymerase chain reaction. RESULTS Oncocytic PDTCs showed a specific immune-related gene expression profile, with higher expression of LAIR2, CD274, DEFB1, IRAK1, CAMP, LCN2, LY96, and APOE, and lower expression of NOD1, as compared to conventional PDTCs. This molecular signature was associated with increased intratumoral lymphocytic infiltration, PD-L1 expression, and adverse outcome. Three of these genes, CD274, DEFB1, and IRAK1, as well as PD-L1 expression, were also the hallmarks of HCCs as compared to FTCs. By contrast, the panel of genes differentially regulated in PDTCs as compared to WDFCs was unrelated to the oncocytic phenotype. CONCLUSIONS Our results revealed a distinctive immune-related gene expression profile of oncocytic PDTC and confirmed a more aggressive outcome in this cancer subtype. These findings may provide guidance when exploring novel immunotherapeutic options for oncocytic PDTC patients.
Collapse
MESH Headings
- Adenocarcinoma, Follicular/genetics
- Adenocarcinoma, Follicular/immunology
- Adenocarcinoma, Follicular/mortality
- Adenocarcinoma, Follicular/pathology
- Adenoma, Oxyphilic/genetics
- Adenoma, Oxyphilic/immunology
- Adenoma, Oxyphilic/mortality
- Adenoma, Oxyphilic/pathology
- Adult
- Aged
- Aged, 80 and over
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunity/genetics
- Male
- Microarray Analysis
- Middle Aged
- Oxyphil Cells/metabolism
- Oxyphil Cells/pathology
- Retrospective Studies
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/immunology
- Thyroid Neoplasms/mortality
- Thyroid Neoplasms/pathology
- Transcriptome
- Tumor Escape/genetics
Collapse
Affiliation(s)
- Jasna Metovic
- Department of Oncology, Pathology Unit of Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Chiara Vignale
- Department of Oncology, Pathology Unit of Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Laura Annaratone
- Department of Medical Sciences, Pathology Unit, University of Turin, Turin, Italy
- Candiolo Cancer Institute, Pathology Division, FPO-IRCCS, Candiolo, Italy
| | - Simona Osella-Abate
- Department of Medical Sciences, Pathology Unit, University of Turin, Turin, Italy
| | - Francesca Maletta
- Department of Oncology, Pathology Unit of Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Ida Rapa
- Department of Oncology, Pathology Unit of San Luigi Hospital, University of Turin, Orbassano, Turin, Italy
| | - Francesco Cabutti
- Department of Oncology, Pathology Unit of Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Silvia Patriarca
- Piedmont Cancer Registry-CRPT, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Marco Gallo
- Department of Medical Sciences, Oncological Endocrinology Unit, Città della Salute e della Scienza Hospital, University of Turin, Turin, Italy
| | - Yuri E Nikiforov
- Department of Pathology, Division of Molecular Genomic Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marco Volante
- Department of Oncology, Pathology Unit of San Luigi Hospital, University of Turin, Orbassano, Turin, Italy
| | - Mauro Papotti
- Department of Oncology, Pathology Unit of Città della Salute e della Scienza, University of Turin, Turin, Italy
| |
Collapse
|
35
|
Iron Dysregulation in Human Cancer: Altered Metabolism, Biomarkers for Diagnosis, Prognosis, Monitoring and Rationale for Therapy. Cancers (Basel) 2020; 12:cancers12123524. [PMID: 33255972 PMCID: PMC7761132 DOI: 10.3390/cancers12123524] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Iron is the more abundant metal ion in humans. It is essential for life as it has a role in various cellular processes involved, for instance, in cell metabolism and DNA synthesis. These functions are crucial for cell proliferation, and it is therefore not surprising that iron is accumulated in tumors. In this review, we describe normal and altered iron homeostasis mechanisms. We also provide a vision of iron-related proteins with altered expression in cancers and discuss their potential as diagnostic and/or prognostic biomarkers. Finally, we give an overview of therapeutic strategies acting on iron metabolism to fight against cancers. Abstract Iron (Fe) is a trace element that plays essential roles in various biological processes such as DNA synthesis and repair, as well as cellular energy production and oxygen transport, and it is currently widely recognized that iron homeostasis is dysregulated in many cancers. Indeed, several iron homeostasis proteins may be responsible for malignant tumor initiation, proliferation, and for the metastatic spread of tumors. A large number of studies demonstrated the potential clinical value of utilizing these deregulated proteins as prognostic and/or predictive biomarkers of malignancy and/or response to anticancer treatments. Additionally, the iron present in cancer cells and the importance of iron in ferroptosis cell death signaling pathways prompted the development of therapeutic strategies against advanced stage or resistant cancers. In this review, we select relevant and promising studies in the field of iron metabolism in cancer research and clinical oncology. Besides this, we discuss some co-existing discrepant findings. We also present and discuss the latest lines of research related to targeting iron, or its regulatory pathways, as potential promising anticancer strategies for human therapy. Iron chelators, such as deferoxamine or iron-oxide-based nanoparticles, which are already tested in clinical trials, alone or in combination with chemotherapy, are also reported.
Collapse
|
36
|
Lipocalin 2 as a potential systemic biomarker for central serous chorioretinopathy. Sci Rep 2020; 10:20175. [PMID: 33214636 PMCID: PMC7677530 DOI: 10.1038/s41598-020-77202-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
No systemic biomarker of Central Serous Chorioretinopathy (CSCR) has been identified. Lipocalin 2 (LCN2 or NGAL), alone or complexed with MMP-9 (NGAL/MMP-9), is increased in several retinal disorders. Serum levels of LCN2 and NGAL/MMP-9 were measured in CSCR patients (n = 147) with chronic (n = 76) or acute/recurrent disease (n = 71) and in age- and sex-matched healthy controls (n = 130). Samples with CRP > 5 mg/L, creatinine > 100 µmol/L, and/or urea > 7.5 mmol/L were excluded. Serum LCN2 was lower in CSCR patients than controls (81.4 ± 48.7 vs 107.3 ± 44.5 ng/ml, p < 0.0001), and lower in acute/recurrent CSCR than controls (p < 0.001) and chronic CSCR (p = 0.006). Serum NGAL/MMP-9 was lower in CSCR patients than controls (47.2 ± 40.7 vs 74.1 ± 42.6, p < 0.0001), and lower in acute/recurrent CSCR than controls (p < 0.001) and chronic CSCR (p = 0.002). A ROC curve showed that for LCN2 serum levels, the 80-ng/ml cutoff value allows to discriminate acute/recurrent CSCR from controls with 80.3% sensitivity and 75.8% specificity, and for NGAL/MMP-9 serum levels, a 38-ng/ml cutoff value allows to discriminate acute/recurrent CSCR from controls with 69.6% sensitivity and 80.3% specificity. In both acute and chronic CSCR, low serum LCN2 and NGAL/MMP-9, provide a biological link between the two CSCR forms, and potential susceptibility to oxidative stress and innate immune dysregulation in CSCR.
Collapse
|
37
|
Yammine L, Zablocki A, Baron W, Terzi F, Gallazzini M. Lipocalin-2 Regulates Epidermal Growth Factor Receptor Intracellular Trafficking. Cell Rep 2020; 29:2067-2077.e6. [PMID: 31722218 DOI: 10.1016/j.celrep.2019.10.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 08/02/2019] [Accepted: 10/03/2019] [Indexed: 11/27/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) activation and lipocalin-2 (Lcn2) expression are frequently observed in the same pathological contexts, such as cancers or chronic kidney disease (CKD). However, the significance of this association is unknown. Here, we describe the role of Lcn2 in regulating EGFR trafficking. We show that Lcn2 increases EGFR cell surface abundance and is required for transforming growth factor α (TGF-α)-induced EGFR recycling to the plasma membrane and sustained activation. Lcn2 binds to the intracellular domain of EGFR in late endosomal compartments and inhibits its lysosomal degradation. Consistently, Lcn2 enhances EGFR-induced cell migration after TGF-α stimulation. In vivo, Lcn2 gene inactivation prevents EGFR recycling to the plasma membrane in an experimental model of CKD. Remarkably, this is associated with a dramatic decrease of renal lesions. Together, our data identify Lcn2 as a key mediator of EGFR trafficking processes. Hence, therapeutic inhibition of Lcn2 may counteract the deleterious effect of EGFR activation.
Collapse
Affiliation(s)
- Lucie Yammine
- Mechanisms and Therapeutic Strategies of Chronic Kidney Disease, INSERM U1151-CNRS UMR 8253, Institut Necker Enfants Malades, Département "Croissance et Signalisation," Hôpital Necker Enfants Malades, Université Paris Descartes, 149 Rue de Sèvres, Paris 75015, France
| | - Aniela Zablocki
- Mechanisms and Therapeutic Strategies of Chronic Kidney Disease, INSERM U1151-CNRS UMR 8253, Institut Necker Enfants Malades, Département "Croissance et Signalisation," Hôpital Necker Enfants Malades, Université Paris Descartes, 149 Rue de Sèvres, Paris 75015, France
| | - William Baron
- Mechanisms and Therapeutic Strategies of Chronic Kidney Disease, INSERM U1151-CNRS UMR 8253, Institut Necker Enfants Malades, Département "Croissance et Signalisation," Hôpital Necker Enfants Malades, Université Paris Descartes, 149 Rue de Sèvres, Paris 75015, France
| | - Fabiola Terzi
- Mechanisms and Therapeutic Strategies of Chronic Kidney Disease, INSERM U1151-CNRS UMR 8253, Institut Necker Enfants Malades, Département "Croissance et Signalisation," Hôpital Necker Enfants Malades, Université Paris Descartes, 149 Rue de Sèvres, Paris 75015, France
| | - Morgan Gallazzini
- Mechanisms and Therapeutic Strategies of Chronic Kidney Disease, INSERM U1151-CNRS UMR 8253, Institut Necker Enfants Malades, Département "Croissance et Signalisation," Hôpital Necker Enfants Malades, Université Paris Descartes, 149 Rue de Sèvres, Paris 75015, France.
| |
Collapse
|
38
|
Jiang M, Qiao M, Zhao C, Deng J, Li X, Zhou C. Targeting ferroptosis for cancer therapy: exploring novel strategies from its mechanisms and role in cancers. Transl Lung Cancer Res 2020; 9:1569-1584. [PMID: 32953528 PMCID: PMC7481593 DOI: 10.21037/tlcr-20-341] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Ferroptosis is a novel form of non-apoptotic regulated cell death (RCD), with distinct characteristics and functions in physical conditions and multiple diseases such as cancers. Unlike apoptosis and autophagy, this new RCD is an iron-dependent cell death with features of lethal accumulation of reactive oxygen species (ROS) and over production of lipid peroxidation. Excessive iron from aberrant iron metabolisms or the maladjustment of the two main redox systems thiols and lipid peroxidation role as the major causes of ROS generation, and the redox-acrive ferrous (intracellular labile iron) is a crucial factor for the lipid peroxidation. Regulation of ferrroptosis also involves different pathways such as mevalonate pathway, P53 pathway and p62-Keap1-Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway. Ferroptosis roles as a double-edged sword either suppressing or promoting tumor progression with the release of multiple signaling molecules in the tumor microenvironment. Emerging evidence suggests ferroptosis as a potential target for cancer therapy and ferroptosis inducers including small molecules and nanomaterials have been developed. The application of ferroptosis inducers also relates to overcoming drug resistance and preventing tumor metastasis, and may become a promising strategy combined with other anti-cancer therapies. Here, we summarize the ferroptosis characters from its underlying basis and role in cancer, followed by its possible applications in cancer therapies and challenges maintained.
Collapse
Affiliation(s)
- Minlin Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Tongji University, Shanghai, China
| | - Meng Qiao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Tongji University, Shanghai, China
| | - Chuanliang Zhao
- Department of Otolaryngology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Juan Deng
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Tongji University, Shanghai, China
| | - Xuefei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
Biological Functions and Therapeutic Potential of Lipocalin 2 in Cancer. Int J Mol Sci 2020; 21:ijms21124365. [PMID: 32575507 PMCID: PMC7352275 DOI: 10.3390/ijms21124365] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/05/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022] Open
Abstract
Lipocalin-2 (LCN2) is a secreted glycoprotein linked to several physiological roles, including transporting hydrophobic ligands across cell membranes, modulating immune responses, maintaining iron homeostasis, and promoting epithelial cell differentiation. Although LNC2 is expressed at low levels in most human tissues, it is abundant in aggressive subtypes of cancer, including breast, pancreas, thyroid, ovarian, colon, and bile duct cancers. High levels of LCN2 have been associated with increased cell proliferation, angiogenesis, cell invasion, and metastasis. Moreover, LCN2 modulates the degradation, allosteric events, and enzymatic activity of matrix metalloprotease-9, a metalloprotease that promotes tumor cell invasion and metastasis. Hence, LCN2 has emerged as a potential therapeutic target against many cancer types. This review summarizes the most relevant findings regarding the expression, biological roles, and regulation of LCN2, as well as the proteins LCN2 interacts with in cancer. We also discuss the approaches to targeting LCN2 for cancer treatment that are currently under investigation, including the use of interference RNAs, antibodies, and gene editing.
Collapse
|
40
|
Brown RAM, Richardson KL, Kabir TD, Trinder D, Ganss R, Leedman PJ. Altered Iron Metabolism and Impact in Cancer Biology, Metastasis, and Immunology. Front Oncol 2020; 10:476. [PMID: 32328462 PMCID: PMC7160331 DOI: 10.3389/fonc.2020.00476] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022] Open
Abstract
Iron is an essential nutrient that plays a complex role in cancer biology. Iron metabolism must be tightly controlled within cells. Whilst fundamental to many cellular processes and required for cell survival, excess labile iron is toxic to cells. Increased iron metabolism is associated with malignant transformation, cancer progression, drug resistance and immune evasion. Depleting intracellular iron stores, either with the use of iron chelating agents or mimicking endogenous regulation mechanisms, such as microRNAs, present attractive therapeutic opportunities, some of which are currently under clinical investigation. Alternatively, iron overload can result in a form of regulated cell death, ferroptosis, which can be activated in cancer cells presenting an alternative anti-cancer strategy. This review focuses on alterations in iron metabolism that enable cancer cells to meet metabolic demands required during different stages of tumorigenesis in relation to metastasis and immune response. The strength of current evidence is considered, gaps in knowledge are highlighted and controversies relating to the role of iron and therapeutic targeting potential are discussed. The key question we address within this review is whether iron modulation represents a useful approach for treating metastatic disease and whether it could be employed in combination with existing targeted drugs and immune-based therapies to enhance their efficacy.
Collapse
Affiliation(s)
- Rikki A. M. Brown
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
- UWA Medical School, University of Western Australia, Perth, WA, Australia
| | - Kirsty L. Richardson
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Tasnuva D. Kabir
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Debbie Trinder
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
- UWA Medical School, University of Western Australia, Perth, WA, Australia
| | - Ruth Ganss
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Peter J. Leedman
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
- UWA Medical School, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
41
|
Gumpper K, Dangel AW, Pita-Grisanti V, Krishna SG, Lara LF, Mace T, Papachristou GI, Conwell DL, Hart PA, Cruz-Monserrate Z. Lipocalin-2 expression and function in pancreatic diseases. Pancreatology 2020; 20:419-424. [PMID: 31932215 PMCID: PMC7160010 DOI: 10.1016/j.pan.2020.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 02/08/2023]
Abstract
Lipocalin-2 (LCN2) is a secreted molecule, expressed in various cell types, that is involved in the progression of numerous diseases and disorders. The biological functions and expression levels of LCN2 in diseases including pancreatic cancer, pancreatitis (acute and chronic), and diabetes mellitus, suggest the potential role of LCN2 as a biomarker and/or therapeutic target. However, findings on the role of LCN2 in pancreatic diseases have been contradictory. In pancreatic cancer and pancreatitis, LCN2 has been identified as a potential biomarker; increased expression levels in various biological specimens correlate with the presence of the disease and may be able to differentiate cancer and chronic pancreatitis from healthy subjects. LCN2 is also known to be an adipokine; it is upregulated in obesity and is a common co-factor in the development of pancreatic diseases. Emerging research suggests LCN2 is elevated in type 2 diabetes mellitus, but the exact role of LCN2 in this disease is not clear. In this review, we summarize research on LCN2 as it relates to pancreatic diseases, highlighting the discrepancies in the literature. By explaining and clarifying the role of LCN2 in these disorders, we aim to promote research in developing novel diagnostic and treatment strategies to reduce the burden of pancreatic diseases.
Collapse
Affiliation(s)
- Kristyn Gumpper
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Andrew William Dangel
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Valentina Pita-Grisanti
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Somashekar G Krishna
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Luis F Lara
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Thomas Mace
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Georgios I Papachristou
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Darwin L Conwell
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Phil A Hart
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
42
|
Lin A, Inman RD, Streutker CJ, Zhang Z, Pritzker KPH, Tsui HW, Tsui FWL. Lipocalin 2 links inflammation and ankylosis in the clinical overlap of inflammatory bowel disease (IBD) and ankylosing spondylitis (AS). Arthritis Res Ther 2020; 22:51. [PMID: 32188494 PMCID: PMC7081573 DOI: 10.1186/s13075-020-02149-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 03/09/2020] [Indexed: 12/16/2022] Open
Abstract
Background Little is known about the mechanisms underlying the clinical overlap between gut inflammation and joint ankylosis, as exemplified by the concurrence of inflammatory bowel diseases (IBD) and ankylosing spondylitis (AS). As dysbiosis may serve as a common contributor, the anti-microbial pleiotropic factor lipocalin 2 could be a potential mediator due to its roles in inflammation and bone homeostasis. Methods Baseline colonic pathology was conducted in the ank/ank mouse model. Serum lipocalin 2 was analyzed by ELISA, in ank/ank mutants versus C3FeB6-A/Aw-jwt/wt, in patients with concurrent AS-IBD, AS alone, IBD alone, or mechanical back pain, and in healthy controls. In the ank/ank mouse model, the expression of nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) was examined by real-time PCR. Intraperitoneal injection was done with the PPARγ agonist rosiglitazone or antagonist bisphenol A diglycidyl ether for four consecutive days. Serum levels of lipocalin 2 were examined on the sixth day. Results This study showed that the ank/ank mice with fully fused spines had concurrent colonic inflammation. By first using the ank/ank mouse model with progressive ankylosis and subclinical colonic inflammation, confirmed in patients with concurrent AS and IBD, elevated circulating lipocalin 2 levels were associated with the coexisting ankylosis and gut inflammation. The intracellular pathway of lipocalin 2 was further investigated with the ank/ank mouse model involving PPARγ. Colonic expression of PPARγ was negatively associated with the degree of gut inflammation. The PPARγ agonist rosiglitazone treatment significantly upregulated the serum levels of lipocalin 2, suggesting a potential regulatory role of PPARγ in the aberrant expression of lipocalin 2. Conclusions In summary, lipocalin 2 modulated by PPARγ could be a potential pathway involved in concurrent inflammation and ankylosis in AS and IBD.
Collapse
Affiliation(s)
- Aifeng Lin
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada. .,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada. .,KeyIntel Medical Inc, Toronto, Ontario, Canada.
| | - Robert D Inman
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Immunology and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Catherine J Streutker
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Li Ka Shing Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Zhenbo Zhang
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kenneth P H Pritzker
- KeyIntel Medical Inc, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Hing Wo Tsui
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Florence W L Tsui
- KeyIntel Medical Inc, Toronto, Ontario, Canada.,Department of Immunology and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Miller I, Schlosser S, Palazzolo L, Veronesi MC, Eberini I, Gianazza E. Some more about dogs: Proteomics of neglected biological fluids. J Proteomics 2020; 218:103724. [PMID: 32126321 DOI: 10.1016/j.jprot.2020.103724] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/28/2020] [Indexed: 01/01/2023]
Abstract
We report in this manuscript what is known about the protein makeup of a selection of biological fluids in the domestic dog. The samples we review - amniotic and allantoic fluid, seminal fluid, saliva, bile, synovial fluid, tears - are still very poorly characterized in this species. For some of them we can present results from our own, mainly unpublished experiments. SIGNIFICANCE: The dog is one of the most widespread companion animals, and also of medical relevance as model species for some human diseases. Still, investigation of body fluids other than serum and urine is not so commonly undertaken, although - like in humans - also these sample types may have potential for diagnostic purposes. We compile published data about proteomes of fetal fluids, seminal plasma, saliva, bile, synovial fluid and tears, enriched by some yet unpublished data of our own (proteins of amniotic and allantoic fluid, tears). Closing gaps in our knowledge on dog proteins will further our understanding of (patho)physiological processes.
Collapse
Affiliation(s)
- Ingrid Miller
- Institut für Medizinische Biochemie, Veterinärmedizinische Universität Wien, Veterinärplatz 1, A-1210 Wien, Austria.
| | - Sarah Schlosser
- VetCore, Veterinärmedizinische Universität Wien, Veterinärplatz 1, A-1210 Wien, Austria
| | - Luca Palazzolo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, I-20133 Milano, Italy
| | - Maria Cristina Veronesi
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Via dell'Università 6, 26900 Lodi, Italy
| | - Ivano Eberini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, I-20133 Milano, Italy
| | - Elisabetta Gianazza
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, I-20133 Milano, Italy
| |
Collapse
|
44
|
Simulated Microgravity Influences VEGF, MAPK, and PAM Signaling in Prostate Cancer Cells. Int J Mol Sci 2020; 21:ijms21041263. [PMID: 32070055 PMCID: PMC7072928 DOI: 10.3390/ijms21041263] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/31/2020] [Accepted: 02/11/2020] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer is one of the leading causes of cancer mortality in men worldwide. An unusual but unique environment for studying tumor cell processes is provided by microgravity, either in space or simulated by ground-based devices like a random positioning machine (RPM). In this study, prostate adenocarcinoma-derived PC-3 cells were cultivated on an RPM for time periods of 3 and 5 days. We investigated the genes associated with the cytoskeleton, focal adhesions, extracellular matrix, growth, survival, angiogenesis, and metastasis. The gene expression of signaling factors of the vascular endothelial growth factor (VEGF), mitogen-activated protein kinase (MAPK), and PI3K/AKT/mTOR (PAM) pathways was investigated using qPCR. We performed immunofluorescence to study the cytoskeleton, histological staining to examine the morphology, and a time-resolved immunofluorometric assay to analyze the cell culture supernatants. When PC-3 cells were exposed to simulated microgravity (s-µg), some cells remained growing as adherent cells (AD), while most cells detached from the cell culture flask bottom and formed multicellular spheroids (MCS). After 3-day RPM exposure, PC-3 cells revealed significant downregulation of the VEGF, SRC1, AKT, MTOR, and COL1A1 gene expression in MCS, whereas FLT1, RAF1, MEK1, ERK1, FAK1, RICTOR, ACTB, TUBB, and TLN1 mRNAs were not significantly changed. ERK2 and TLN1 were elevated in AD, and FLK1, LAMA3, COL4A5, FN1, VCL, CDH1, and NGAL mRNAs were significantly upregulated in AD and MCS after 3 days. After a 5-day culture in s-µg, the PC-3 cells showed significant downregulations of VEGF mRNA in AD and MCS, and FN1, CDH1, and LAMA3 in AD and SCR1 in MCS. In addition, we measured significant upregulations in FLT1, AKT, ERK1, ERK2, LCN2, COL1A1, TUBB, and VCL mRNAs in AD and MCS, and increases in FLK1, FN1, and COL4A5 in MCS as well as LAMB2, CDH1, RAF1, MEK1, SRC1, and MTOR mRNAs in AD. FAK1 and RICTOR were not altered by s-µg. In parallel, the secretion rate of VEGFA and NGAL proteins decreased. Cytoskeletal alterations (F-actin) were visible, as well as a deposition of collagen in the MCS. In conclusion, RPM-exposure of PC-3 cells induced changes in their morphology, cytoskeleton, and extracellular matrix protein synthesis, as well as in their focal adhesion complex and growth behavior. The significant upregulation of genes belonging to the PAM pathway indicated their involvement in the cellular changes occurring in microgravity.
Collapse
|
45
|
Implication and role of neutrophil gelatinase-associated lipocalin in cancer: lipocalin-2 as a potential novel emerging comprehensive therapeutic target for a variety of cancer types. Mol Biol Rep 2020; 47:2327-2346. [PMID: 31970626 DOI: 10.1007/s11033-020-05261-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/16/2020] [Indexed: 12/18/2022]
Abstract
Cancer is a leading cause of mortalities worldwide. Over the past few decades, exploration of molecular mechanisms behind cancer initiation and progression has been of great interest in the viewpoint of both basic and clinical scientists. It is generally believed that identification of key molecules implicated in cancer pathology not only improves our understanding of the disease, but also could result in introduction of novel therapeutic strategies. Neutrophil gelatinase-associated lipocalin (NGAL)/lipocalin-2 (LCN2) is a member of lipocalin superfamily with a variety of functions. Although the main function of LCN2 is still unknown, many studies confirmed its significant role in the initiation, progression, and metastasis of various types of cancer. Furthermore, aberrant expression of LCN2 is also concerned with the chemo- and radio-resistant phenotypes of tumors. Here, we will review the contribution of known functions of LCN2 to the pathophysiology of cancer. We also highlight how the deregulated expression of LCN2 is associated with a variety of fatal types of cancer for which there are no effective therapeutic modalities. The unique and multiple functions of LCN2 and its widespread expression in different types of cancer prompted us to suggest LCN2 could be considered either as a valuable diagnostic and prognostic biomarker or as a potential novel therapeutic target.
Collapse
|
46
|
Huang X, Slavkovic S, Song E, Botta A, Mehrazma B, Lento C, Johnson PE, Sweeney G, Wilson DJ. A Unique Conformational Distortion Mechanism Drives Lipocalin 2 Binding to Bacterial Siderophores. ACS Chem Biol 2020; 15:234-242. [PMID: 31613081 DOI: 10.1021/acschembio.9b00820] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Lcn2 is a host defense protein induced via the innate immune response to sequester iron-loaded bacterial siderophores. However, excess or prolonged elevation of Lcn2 levels can induce adverse cellular effects, including oxidative stress and inflammation. In this work, we use Hydrogen-Deuterium eXchange (HDX) and Isothermal Titration Calorimetry (ITC) to characterize the binding interaction between Lcn2 and siderophores enterobactin and 2,3-DHBA, in the presence and absence of iron. Our results indicate a rare "Type II" interaction in which binding of siderophores drives the protein conformational equilibrium toward an unfolded state. Linking our molecular model to cellular assays, we demonstrate that this "distorted binding mode" facilitates a deleterious cellular accumulation of reactive oxygen species that could represent the molecular origin of Lcn2 pathology. These results add important insights into mechanisms of Lcn2 action and have implications in Lcn2-mediated effects including inflammation.
Collapse
|
47
|
Abstract
Mass spectrometry-based proteomics analysis could categorize proteins and study their interactions in large scale in human cancers. By this method, many proteins are upregulated or downregulated in esophageal squamous cell carcinoma (ESCC) when compared to nonneoplastic esophageal mucosae. The method can also be used to identify novel, effective biomarkers for early diagnosis or predict prognosis of patients with ESCC. These changes are associated with different clinical and pathological parameters. Different biological matrices such as pathological tissue, body fluids, and cancer cell lines-based proteomics have widely been used. Herein, we described cell line-based label-free shotgun proteomics (in-solution tryptic digestion) to identify the protein biomarkers differently expressed in ESCC.
Collapse
|
48
|
Swaminathan G, Krishnamurthy VK, Sridhar S, Robson DC, Ning Y, Grande-Allen KJ. Hypoxia Stimulates Synthesis of Neutrophil Gelatinase-Associated Lipocalin in Aortic Valve Disease. Front Cardiovasc Med 2019; 6:156. [PMID: 31737648 PMCID: PMC6828964 DOI: 10.3389/fcvm.2019.00156] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022] Open
Abstract
Objective: Aortic valve disease is commonly found in the elderly population. It is characterized by dysregulated extracellular matrix remodeling followed by extensive microcalcification of the aortic valve and activation of valve interstitial cells. The mechanism behind these events are largely unknown. Studies have reported expression of hypoxia inducible factor-1 alpha (HIF1α) in calcific nodules in aortic valve disease, therefore we investigated the effect of hypoxia on extracellular matrix remodeling in aged aortic valves. Approach and Results: Western blotting revealed elevated expression of HIF1α and the complex of matrix metalloprotease 9 (MMP9) and neutrophil gelatinase-associated lipocalin (NGAL) in aged porcine aortic valves cultured under hypoxic conditions. Consistently, immunofluorescence staining showed co-expression of MMP9 and NGAL in the fibrosa layer of these porcine hypoxic aortic valves. Gelatinase zymography demonstrated that the activity of MMP9-NGAL complex was significantly increased in aortic valves in 13% O2 compared to 20% O2. Importantly, the presence of ectopic elastic fibers in the fibrosa of hypoxic aortic valves, also detected in human diseased aortic valves, suggests altered elastin homeostasis due to hypoxia. Conclusion: This study demonstrates that hypoxia stimulates pathological extracellular matrix remodeling via expression of NGAL and MMP9 by valve interstitial cells.
Collapse
|
49
|
Chen YC, Chang SC, Huang YH, Lee YJ, Chang CC, Liao JW, Hsu WL. Expression and the molecular forms of neutrophil gelatinase-associated lipocalin and matrix metalloproteinase 9 in canine mammary tumours. Vet Comp Oncol 2019; 17:427-438. [PMID: 31050171 DOI: 10.1111/vco.12488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/10/2019] [Accepted: 04/26/2019] [Indexed: 01/01/2023]
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL) is a new biomarker for renal injury. It is also involved in tumorigenesis of different human cancer types. The oncogenic role of NGAL is related to its molecular forms, and heterodimer formation with matrix metalloproteinase 9 (MMP9) promotes human breast cancer (HBC) invasion and metastasis. To date, the levels of NGAL and NGAL/MMP9 complex have not yet been explored in canine mammary tumours (CMTs). Hence, this study aimed to investigate whether NGAL and its molecular forms could be the biomarker for CMT diagnosis. To this end, expression profile of NGAL and MMP9 in mammary epithelial cells as well as in urine samples were detected. By immunohistochemistry staining, NGAL was expressed at variable levels. Unlike HBC, a significant reduction in NGAL expression was demonstrated in benign and malignant CMTs as compared with normal controls. Additionally, NGAL expression was significantly reduced in dogs with metastatic CMTs. By contrast, the mean score of MMP9 expression in ascending order was normal groups, benign, and malignant CMTs. Interestingly, analysis of the molecular form revealed the NGAL/MMP9 complex presents in most mammary tissues and urine of dogs with benign or malignant CMTs, whereas the complex was absent in samples from dogs without CMTs. In conclusion, NGAL and MMP9 are ubiquitously expressed in canine mammary epithelial cells in normal and cancerous status. However, the NGAL/MMP9 complex exclusively presents in mammary tissues and urine of dogs with tumours.
Collapse
Affiliation(s)
- Yi-Chen Chen
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Shih-Chieh Chang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan.,Veterinary Medical Teaching Hospital, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Han Huang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Ya-Jane Lee
- Institute of Veterinary Clinical Science, School of Veterinary Medicine, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Chao-Chin Chang
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Wei-Li Hsu
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
50
|
Montalbo R, Lozano JJ, Izquierdo L, Ingelmo-Torres M, BaÑos C, Palou J, Van der Heijden AG, Medina R, Schmidbauer J, Prat A, Ribal MJ, Alcaraz A, Mengual L. Ability of a urine gene expression classifier to reduce the number of follow-up cystoscopies in bladder cancer patients. Transl Res 2019; 208:73-84. [PMID: 30771285 DOI: 10.1016/j.trsl.2019.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/18/2019] [Accepted: 02/04/2019] [Indexed: 11/28/2022]
Abstract
This study aimed to improve our previous urine gene expression classifiers focusing on the detection of non-high-risk non-muscle-invasive bladder cancer (NMIBC), and develop a new classifier able to decrease the frequency of cystoscopies during bladder cancer (BC) patients' surveillance. A total of 597 urines from BC patients, controls and patients in follow-up for BC (PFBC) were included. The study has 3 phases. In the urinary biomarker discovery phase, 84 urines from BC and control patients were retrospectively included and analyzed by Ribonucleic Acid (RNA) sequencing. In the classifier development phase, a total of 132 selected genes from previous phase were evaluated by nCounter in 214 prospectively collected urines from PFBC (98 with tumor). A diagnostic classifier was generated by logistic regression. Finally, in the classifier validation phase, a multicentric and international cohort of 248 urines (134 BC and 114 nonrecurrent PFBC) was used to validate classifier performance. A total of 521 genes were found differentially expressed between non-high-risk NMIBC samples and all other groups (P < 0.05). An 8-gene diagnostic classifier with an area under curve (AUC) of 0.893 was developed. Validation of this classifier in a cohort of PFBC achieved an overall sensitivity (SN) and a negative predictive value (NPV) of 96% and 97%, respectively (AUC = 0.823). Notably, this accuracy was maintained in non-high-risk NMIBC group (SN = 94%; NPV = 98%). In conclusion, this 8-gene expression classifier has high SN and NPV in a real clinical scenario. The use of this classifier can reduce the number of follow-up cystoscopies in PFBC, although assessing its final place in clinical setting is necessary.
Collapse
Affiliation(s)
- Ruth Montalbo
- Department and Laboratory of Urology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centre de Recerca Biomèdica CELLEX, Universitat de Barcelona, Barcelona, Spain
| | | | - Laura Izquierdo
- Department and Laboratory of Urology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centre de Recerca Biomèdica CELLEX, Universitat de Barcelona, Barcelona, Spain
| | - Mercedes Ingelmo-Torres
- Department and Laboratory of Urology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centre de Recerca Biomèdica CELLEX, Universitat de Barcelona, Barcelona, Spain
| | - Carmen BaÑos
- Department and Laboratory of Urology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centre de Recerca Biomèdica CELLEX, Universitat de Barcelona, Barcelona, Spain
| | - Joan Palou
- Department of Urology, Fundació Puigvert, Barcelona, Spain
| | | | - Rafael Medina
- Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Joerg Schmidbauer
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Aleix Prat
- Oncology Department, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Maria J Ribal
- Department and Laboratory of Urology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centre de Recerca Biomèdica CELLEX, Universitat de Barcelona, Barcelona, Spain
| | - Antonio Alcaraz
- Department and Laboratory of Urology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centre de Recerca Biomèdica CELLEX, Universitat de Barcelona, Barcelona, Spain
| | - Lourdes Mengual
- Department and Laboratory of Urology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centre de Recerca Biomèdica CELLEX, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|