1
|
Liu H, Mei M, Lin S, Luo J, Huang S, Zhou J. Wuling San regulates AVPR2-cAMP-PKA-CREB pathway to delay cellular senescence and ameliorate acute kidney injury. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119679. [PMID: 40216046 DOI: 10.1016/j.jep.2025.119679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/17/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cellular senescence in renal resident cells plays a pivotal role in the progression of acute kidney injury (AKI), necessitating the expansion of effective drug targets. Traditional Chinese medicine (TCM) formulations, characterized by their multi-target effects, offer a promising perspective for advancing research on AKI. Wuling San (WLS), a well-established compound used in treating urological disorders, has yet to elucidate its potential pharmacological targets and mechanisms in ameliorating AKI and delaying cellular senescence. AIM OF THE STUDY This study sought to elucidate the mechanisms by which WLS modulates the AVPR2-cAMP-PKA-CREB pathway to mitigate cellular senescence and promote recovery from AKI. METHODS We first prepared WLS-containing serum and performed RT-qPCR experiments to screen for GPCRs that were differentially expressed in response to WLS. Next, we established an in vitro AKI mouse model to assess the renal protective effects of the WLS by measuring renal function, renal pathology, and oxidative stress levels. After this, we performed RNA sequencing (RNA-Seq) profiling to identify differentially expressed genes (DEGs) affected by WLS treatment. We also conducted Gene Ontology (GO) functional enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses to identify potential signaling pathways involved. We then utilized the Gene Expression Omnibus (GEO) data to screen for cellular senescence related differentially expressed genes (CSRDEGs) in AKI patients and performed enrichment analysis, as well as a joint analysis of specific genes in relation to the RNA-Seq profiling results. We also examined how WLS affects the expression of proteins linked to cellular senescence in the AKI mouse model by targeting the AVPR2-cAMP-PKA-CREB pathway. RESULTS WLS markedly enhanced the expression of Arginine Vasopressin Receptor 2 (AVPR2) and ameliorated renal function indicators, as well as pathological changes and oxidative stress levels in the mouse model of AKI. RNA-Seq profiling revealed significant enrichment of the cAMP signaling pathway following WLS intervention. Bioinformatics analysis indicated that genes associated with cellular senescence in AKI patients were notably enriched in the p53 signaling pathway. Data mining from the GEO database, in conjunction with RNA-Seq profiling, demonstrated a substantial reduction in key genes after WLS treatment. Additionally, WLS elevated both the expression and phosphorylation of pivotal proteins within the AVPR2-cAMP-PKA-CREB pathway, while concurrently decreasing proteins associated with cellular senescence. CONCLUSION The results demonstrated that WLS significantly elevated the expression of AVPR2, which may underlie its nephroprotective effects and facilitate the mitigation of AKI by modulating the AVPR2-cAMP-PKA-CREB pathway, ultimately contributing to a delay in cellular senescence.
Collapse
Affiliation(s)
- Hong Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Manxue Mei
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Shuyin Lin
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Jiahui Luo
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Sirong Huang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Jiuyao Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China.
| |
Collapse
|
2
|
Herbstein F, Rosmino J, Guitelman MA, Cagliero J, Gonilski‐Pacin D, Ciancio Del Giudice N, Fiz M, Fuertes M, Arzt E. New insights in cellular senescence: The pituitary model. J Neuroendocrinol 2025; 37:e70008. [PMID: 40032281 PMCID: PMC12045676 DOI: 10.1111/jne.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/06/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
Pituitary tumors are characterized by slow proliferation rates and a high prevalence within the population. The pathogenesis of these tumors remains incompletely understood, although accumulating evidence suggests that the activation of the cellular senescence program, triggered by various stressors and functioning as a brake on cellular proliferation, may contribute to their typically benign nature. Multiple mediators of the senescence response are implicated in this process. Interleukin-6 (IL-6), a proinflammatory cytokine, plays a dual role in pituitary tumor biology. It is involved in both physiological pituitary growth and the senescence-associated secretory phenotype (SASP), where it mediates paracrine-proliferative signals. In addition to its secretory functions, IL-6 has been implicated in the regulation of pituitary senescence through non-secretory mechanisms. Other factors, such as growth hormone (GH), the pituitary tumor-transforming gene (PTTG), and interactions within the tumor microenvironment, including immune cell dynamics, also contribute to the senescence observed in these tumors. This review examines the latest evidence concerning the role of senescence in pituitary tumors, with a particular focus on the contribution of IL-6 to this process.
Collapse
Affiliation(s)
- Florencia Herbstein
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck SocietyBuenos AiresArgentina
| | - Josefina Rosmino
- División EndocrinologíaHospital General de Agudos “Carlos G. Durand”Buenos AiresArgentina
| | | | - Joaquina Cagliero
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck SocietyBuenos AiresArgentina
| | - David Gonilski‐Pacin
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck SocietyBuenos AiresArgentina
| | - Nicolas Ciancio Del Giudice
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck SocietyBuenos AiresArgentina
| | - Manuel Fiz
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck SocietyBuenos AiresArgentina
| | - Mariana Fuertes
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck SocietyBuenos AiresArgentina
- Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y NaturalesUniversidad de Buenos AiresBuenos AiresArgentina
| | - Eduardo Arzt
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) – CONICET – Partner Institute of the Max Planck SocietyBuenos AiresArgentina
- Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y NaturalesUniversidad de Buenos AiresBuenos AiresArgentina
| |
Collapse
|
3
|
Joshi N, Mueller S, Kline C. Current clinical trials for craniopharyngiomas: what's on the horizon? J Neurooncol 2025; 172:281-288. [PMID: 40042714 PMCID: PMC11937174 DOI: 10.1007/s11060-024-04899-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 11/23/2024] [Indexed: 03/26/2025]
Abstract
Craniopharyngiomas are histologically low-grade tumors in the sellar/suprasellar region that grow close to critical structures including the hypothalamus, pituitary gland, and optic chiasm. Due to this challenging location, many patients face long-term complications including neuroendocrine, neurologic, and visual deficits. As a result, there is interest in developing risk-optimized treatments that minimize damage to adjacent normal tissue and limit chronic complications patients face. In recent years, numerous multi-omic characterizations of craniopharyngioma have identified potential targetable markers of craniopharyngioma. In adamantinomatous craniopharyngioma, numerous clinical trials to explore MEK, PD-1, WNT, and IL-6 inhibition are currently active. In papillary craniopharyngioma, targeting BRAF-V600E and MEK with monotherapy and combined therapies are currently being investigated. Further combining of these therapies with radiation and surgical techniques have potential to change existing treatment paradigms and improve the long-term outcome for patients with craniopharyngioma. With our advanced understanding, clinical investigations that target identified oncogenic drivers of craniopharyngioma should continue to center on therapy options that minimize complications faced by patients with this chronic, high morbidity disease.
Collapse
Affiliation(s)
- Nikhil Joshi
- Division of Oncology, Department of Pediatrics, Perelman School of Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sabine Mueller
- Departments of Neurology, Neurological Surgery, and Pediatrics, University of California, San Francisco, CA, 94158, USA.
- Department of Pediatrics, University of Zurich, Zurich, Switzerland.
| | - Cassie Kline
- Division of Oncology, Department of Pediatrics, Perelman School of Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
4
|
Ren Q, Fu J, Wu B, Xie S, Tang B, Hong T, Wu J. Finger-like protrusions in adamantinomatous craniopharyngiomas: implications for recurrence and prognosis following complete tumor resection and predictive factors. Neurosurg Rev 2025; 48:242. [PMID: 39954160 DOI: 10.1007/s10143-025-03412-5] [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: 11/25/2024] [Revised: 01/19/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Finger-like protrusions (FPs) extending into the hypothalamus in adamantinomatous craniopharyngiomas (ACPs) pose surgical challenges and their link to tumor recurrence and patient prognosis following gross total resection (GTR) is unclear. This study aims to determine the association between FPs, tumor recurrence, and prognosis after GTR, while identifying predictors for the presence of FPs. A total of 149 ACP patients undergoing GTR at our institution between January 2015 and December 2023 were retrospectively analyzed and were categorized into FP and non-FP groups based on the histology of the interface between ACP and the hypothalamus. Various parameters were compared, and logistic regression analyses were conducted to identify predictors for the presence of FPs in ACPs. The FP group (n = 50) had higher rates of meningitis (p = 0.020), postoperative hydrocephalus (p = 0.014), pneumonia (p < 0.001), perioperative mortality (p = 0.036), radiological hypothalamic injury (p < 0.001), and hypothalamic dysfunction (p < 0.001) during follow-up. They also showed inferior recurrence-free survival (p = 0.012) and overall survival (p = 0.023). Predictors for FPs in ACPs included solid tumor (OR 5.28, 95% CI 1.81-15.40; p = 0.002) and preoperative MRI without an upward eagle sign (OR 27.65, 95% CI 8.93-85.65; p < 0.001). FPs in ACPs pose challenges for achieving safe radical tumor resection and are linked to higher recurrence rates and unfavorable patient outcomes after GTR. Solid tumor consistency and preoperative MRI without an upward eagle sign could predict the presence of FPs in ACPs involving the hypothalamus. For ACPs demonstrating these characteristics, a more conservative resection should be considered. Additionally, rigorous postoperative monitoring for tumor recurrence is essential for ACPs with FPs, even after GTR.
Collapse
Affiliation(s)
- Quan Ren
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Department of Neurosurgery, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Jun Fu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Department of Neurosurgery, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, No. 90, Bayi Road, Xihu District, Nanchang, Jiangxi, China
| | - Bowen Wu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shenhao Xie
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Bin Tang
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Tao Hong
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Jie Wu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
5
|
Shatara M, Abdelbaki MS. Pediatric Suprasellar Tumors: Unveiling the Mysteries of Craniopharyngioma and Germ Cell Tumors-Insights From Diagnosis to Advanced Therapeutics. Pediatr Neurol 2025; 162:55-68. [PMID: 39561686 DOI: 10.1016/j.pediatrneurol.2024.10.016] [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: 01/21/2024] [Revised: 08/07/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Pediatric suprasellar tumors represent a unique and intricate challenge in the landscape of pediatric neuro-oncology. METHODS We conducted an in-depth literature review, focusing on large clinical trials and major publications in pediatric suprasellar tumors, particularly craniopharyngiomas and germ cell tumors, to provide a comprehensive perspective on the challenges in the diagnosis, treatment, and molecular aspects of these tumors. RESULTS Nestled within the critical confines of the suprasellar region, these tumors manifest against the backdrop of crucial growth and developmental processes. The suprasellar region, housing the pituitary gland and surrounding structures, plays a pivotal role in orchestrating hormonal regulation and growth. The emergence of tumors within this delicate terrain introduces a complex array of challenges, encompassing neurological, endocrinological, and developmental dimensions from damage to the hypothalamic-pituitary axis. CONCLUSIONS This article provides a thorough exploration of pediatric craniopharyngiomas and germ cell tumors, elucidating their clinical presentations, treatment modalities, and outcomes. The focused analysis aims to deepen our understanding of these tumors by offering insights for refined clinical management and improved patient outcomes.
Collapse
Affiliation(s)
- Margaret Shatara
- Department of Pediatric Hematology and Oncology, Children's Hospitals and Clinics of Minnesota, Minneapolis, Minnesota.
| | - Mohamed S Abdelbaki
- The Division of Hematology and Oncology, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
6
|
McHugh D, Durán I, Gil J. Senescence as a therapeutic target in cancer and age-related diseases. Nat Rev Drug Discov 2025; 24:57-71. [PMID: 39548312 DOI: 10.1038/s41573-024-01074-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 11/17/2024]
Abstract
Cellular senescence is a stress response that restrains the growth of aged, damaged or abnormal cells. Thus, senescence has a crucial role in development, tissue maintenance and cancer prevention. However, lingering senescent cells fuel chronic inflammation through the acquisition of a senescence-associated secretory phenotype (SASP), which contributes to cancer and age-related tissue dysfunction. Recent progress in understanding senescence has spurred interest in the development of approaches to target senescent cells, known as senotherapies. In this Review, we evaluate the status of various types of senotherapies, including senolytics that eliminate senescent cells, senomorphics that suppress the SASP, interventions that mitigate senescence and strategies that harness the immune system to clear senescent cells. We also summarize how these approaches can be combined with cancer therapies, and we discuss the challenges and opportunities in moving senotherapies into clinical practice. Such therapies have the potential to address root causes of age-related diseases and thus open new avenues for preventive therapies and treating multimorbidities.
Collapse
Affiliation(s)
- Domhnall McHugh
- Senescence Group, MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Imanol Durán
- Senescence Group, MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Jesús Gil
- Senescence Group, MRC Laboratory of Medical Sciences (LMS), London, UK.
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
7
|
Alcaide C, Perez F, Esteban F, Muñoz M. Substance P and Neurokinin-1 receptor are overexpressed in adamantinomatous craniopharyngioma than in the pituitary gland. Pituitary 2024; 28:5. [PMID: 39724307 DOI: 10.1007/s11102-024-01490-0] [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] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Human adamantinomatous craniopharyngioma (ACP) is a brain tumor that originates at the base of the skull and shows aggressive local behavior, invading sensitive structures such as the optic pathways and hypothalamus. The conventional treatment of the tumor has been surgery and radiotherapy with the consequent development of serious sequelae. It is well known that Substance P (SP) peptide and Neurokinin-1 receptor (NK-1R) are involved in inflammation and cancer progression and its blockage with NK-1R antagonists has been shown to effectively counteract tumor development in preclinical trials. The oncogenic mechanism underlying ACP is based on a secretory phenotype associated with the production of paracrine biomarkers that establish an inflammatory and angiogenic microenvironment for the progression of ACP. METHODS With the aim of describing the existence and distribution of SP/NK-1R in the ACP, we studied by immunohistochemistry the expression of SP and NK-1R in 43 human ACP and compared with healthy pituitary gland samples. RESULTS SP and the NK-1R were overexpressed in all ACP more than in pituitary glands samples. SP expression is found widespread the ACP and is preferentially localized in the nucleus than in cytoplasm of tumor cells. Likewise, areas of glial reaction and endothelial cells also express SP preferentially in the cell nuclei. NK-1R is expressed mainly in the glial reaction, especially in the nuclei and membranes of its inflammatory cells and less prominently in the cytoplasm. In ACP neovessels, NK-1R is expressed in endothelial cells and fibroblasts that constitute their basement membranes. Tumor cells did not show significant NK-1R expression. CONCLUSIONS These findings, reported here for the first time, suggest a role for SP and NK-1R in pituitary gland and ACP and opens the door to future clinical trials on treatment with NK-1R antagonist drugs in ACP patients.
Collapse
Affiliation(s)
- Carlos Alcaide
- Department of Pediatric Oncology, Hospital Regional Universitario de Málaga, Malaga, Spain.
| | - Francisco Perez
- Department of Pathology, Facultad de Medicina, Universidad Camilo José Cela / HM Hospitales, Madrid, Spain
| | - Francisco Esteban
- Department of Otorinolaringology, Facultad de Medicina, Universidad de Sevilla / Hospital Universitario Virgen Del Rocío, Seville, Spain
| | - Miguel Muñoz
- Research Laboratory on Neuropeptides, Institute of Biomedicine of Seville (IBIS), Seville, Spain
| |
Collapse
|
8
|
Chen Y, Wang Z, Huang Q, Wang Y, Yan F, Xiang S, Xu L, Chen Y, Liu X, Chen G, Li M, Zhou Y. Differential proteomic profiles of exosomes in pediatric and adult adamantinomatous craniopharyngioma cyst fluid. Mol Biol Rep 2024; 51:1126. [PMID: 39505756 DOI: 10.1007/s11033-024-10073-y] [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: 08/14/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Adamantinomatous craniopharyngiomas (ACPs), commonly seen in pediatrics and adults often present with large cystic cavities that can compress surrounding tissues, causing severe visual and endocrine symptoms. Complete resection of cystic ACP is challenging, frequently leading to postoperative recurrence. The composition of the cystic fluid is complex, and to date, there has been limited research focusing on exosomes within ACP cyst fluid. METHODS We collected cyst fluid from 12 ACP patients and confirmed the presence of exosomes. Subsequently, we conducted exosomal proteomic analysis using LC-MS/MS. The patients were divided into pediatric and adult groups for the analysis of differential protein enrichment, followed by comprehensive bioinformatics analysis, including GO analysis, KEGG analysis, and PPI network analysis, among other functional pathway and protein interaction analyses. Immunohistochemistry was used to determine the tissue expression distribution of the differential protein APOA1. RESULTS In our data analysis, 64 significantly differentially expressed proteins were identified, with 37 being overexpressed in the pediatric group and 27 in the adult group. Our results revealed that exosomal proteins in the pediatric group were predominantly enriched in modules and pathways related to high-density lipoprotein particle, apolipoprotein receptor binding, and the PPAR signaling pathway. Additionally, APOA1, as the hub protein with the highest connectivity in the differential protein interaction network, may play a critical role in β-amyloid metabolism pathways in pediatric ACP. CONCLUSION This study is the first to construct a proteomic map of ACP cyst fluid exosomes, suggesting significant differences in the tumor microenvironment's lipid metabolism between pediatrics and adults.
Collapse
Affiliation(s)
- Yiguang Chen
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Ziyu Wang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
| | - Qin Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Yaming Wang
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Feng Yan
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Sishi Xiang
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Lixin Xu
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institute, 10005, Stockholm, Sweden.
| | - Xiaohai Liu
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Ge Chen
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Mingchu Li
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Yiqiang Zhou
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
9
|
Marrero-Gutiérrez J, Bueno AC, Martins CS, Coeli-Lacchini FB, Silva-Júnior RMP, Marques Gonçalves GH, Ozaki JGO, de Almeida E Silva DC, Wildemberg LE, da Silva Antunes XL, Dos Santos AC, Machado HR, Santos MV, Moreira AC, Gadelha MR, Vêncio RZN, Antonini SRR, de Castro M. Epigenetic Control of Adamantinomatous Craniopharyngiomas. J Clin Endocrinol Metab 2024; 109:e1867-e1880. [PMID: 38181427 DOI: 10.1210/clinem/dgae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 01/07/2024]
Abstract
INTRODUCTION Studies addressing the methylation pattern in adamantinomatous craniopharyngioma (ACP) are lacking. OBJECTIVE To identify methylation signatures in ACPs regarding clinical presentation and outcome. METHODS Clinical and pathology data were collected from 35 patients with ACP (54% male; 18.1 years [2-68]). CTNNB1 mutations and methylation profile (MethylationEPIC/Array-Illumina) were analyzed in tumoral DNA. Unsupervised machine learning analysis of this comprehensive methylome sample was achieved using hierarchical clustering and multidimensional scaling. Statistical associations between clusters and clinical features were achieved using the Fisher test and global biological process interpretations were aided by Gene Ontology enrichment analyses. RESULTS Two clusters were revealed consistently by all unsupervised methods (ACP-1: n = 18; ACP-2: n = 17) with strong bootstrap statistical support. ACP-2 was enriched by CTNNB1 mutations (100% vs 56%, P = .0006), hypomethylated in CpG island, non-CpG Island sites, and globally (P < .001), and associated with greater tumor size (24.1 vs 9.5 cm3, P = .04). Enrichment analysis highlighted pathways on signaling transduction, transmembrane receptor, development of anatomical structures, cell adhesion, cytoskeleton organization, and cytokine binding, and cell type-specific biological processes as regulation of oligodendrocytes, keratinocyte, and epithelial cells differentiation. CONCLUSION Two clusters of patients with ACP were consistently revealed by unsupervised machine learning methods, with one of them significantly hypomethylated, enriched by CTNNB1 mutated ACPs, and associated with increased tumor size. Enrichment analysis reinforced pathways involved in tumor proliferation and in cell-specific tumoral microenvironment.
Collapse
Affiliation(s)
- Junier Marrero-Gutiérrez
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| | - Ana Carolina Bueno
- Department of Pediatrics, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| | - Clarissa Silva Martins
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| | | | - Rui M Patrício Silva-Júnior
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| | | | - Jorge Guilherme Okanobo Ozaki
- Department of Medical Imaging, Hematology and Oncology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| | - Danillo C de Almeida E Silva
- Department of Computation and Mathematics Biology, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, University of São Paulo, Ribeirao Preto, SP, 14040-901, Brazil
| | - Luiz Eduardo Wildemberg
- Neuroendocrinology Research Center/Endocrinology Section, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro, 21941-913, Brazil
| | - Ximene Lima da Silva Antunes
- Neuroendocrinology Research Center/Endocrinology Section, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro, 21941-913, Brazil
| | - Antônio Carlos Dos Santos
- Department of Medical Imaging, Hematology and Oncology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| | - Helio Rubens Machado
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| | - Marcelo Volpon Santos
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| | - Ayrton Custodio Moreira
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| | - Monica R Gadelha
- Neuroendocrinology Research Center/Endocrinology Section, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro, 21941-913, Brazil
| | - Ricardo Zorzetto Nicoliello Vêncio
- Department of Computation and Mathematics Biology, Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, University of São Paulo, Ribeirao Preto, SP, 14040-901, Brazil
| | - Sonir Roberto R Antonini
- Department of Pediatrics, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| | - Margaret de Castro
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, SP, 14049-900, Brazil
| |
Collapse
|
10
|
Suda M, Paul KH, Tripathi U, Minamino T, Tchkonia T, Kirkland JL. Targeting Cell Senescence and Senolytics: Novel Interventions for Age-Related Endocrine Dysfunction. Endocr Rev 2024; 45:655-675. [PMID: 38500373 PMCID: PMC11405506 DOI: 10.1210/endrev/bnae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/11/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
Multiple changes occur in hormonal regulation with aging and across various endocrine organs. These changes are associated with multiple age-related disorders and diseases. A better understanding of responsible underling biological mechanisms could help in the management of multiple endocrine disorders over and above hormone replacement therapy (HRT). Cellular senescence is involved in multiple biological aging processes and pathologies common in elderly individuals. Cellular senescence, which occurs in many older individuals but also across the lifespan in association with tissue damage, acute and chronic diseases, certain drugs, and genetic syndromes, may contribute to such endocrine disorders as osteoporosis, metabolic syndrome, and type 2 diabetes mellitus. Drugs that selectively induce senescent cell removal, "senolytics,", and drugs that attenuate the tissue-destructive secretory state of certain senescent cells, "senomorphics," appear to delay the onset of or alleviate multiple diseases, including but not limited to endocrine disorders such as diabetes, complications of obesity, age-related osteoporosis, and cancers as well as atherosclerosis, chronic kidney disease, neurodegenerative disorders, and many others. More than 30 clinical trials of senolytic and senomorphic agents have already been completed, are underway, or are planned for a variety of indications. Targeting senescent cells is a novel strategy that is distinct from conventional therapies such as HRT, and thus might address unmet medical needs and can potentially amplify effects of established endocrine drug regimens, perhaps allowing for dose decreases and reducing side effects.
Collapse
Affiliation(s)
- Masayoshi Suda
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Karl H Paul
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| | - Utkarsh Tripathi
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan
| | - Tamara Tchkonia
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - James L Kirkland
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
11
|
Feng H, Li J, Wang H, Wei Z, Feng S. Senescence- and Immunity-Related Changes in the Central Nervous System: A Comprehensive Review. Aging Dis 2024:AD.2024.0755. [PMID: 39325939 DOI: 10.14336/ad.2024.0755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Senescence is a cellular state characterized by an irreversible halt in the cell cycle, accompanied by alterations in cell morphology, function, and secretion. Senescent cells release a plethora of inflammatory and growth factors, extracellular matrix proteins, and other bioactive substances, collectively known as the senescence-associated secretory phenotype (SASP). These excreted substances serve as crucial mediators of senescent tissues, while the secretion of SASP by senescent neurons and glial cells in the central nervous system modulates the activity of immune cells. Senescent immune cells also influence the physiological activities of various cells in the central nervous system. Further, the interaction between cellular senescence and immune regulation collectively affects the physiological and pathological processes of the central nervous system. Herein, we explore the role of senescence in the physiological and pathological processes underlying embryonic development, aging, degeneration, and injury of the central nervous system, through the immune response. Further, we elucidate the role of senescence in the physiological and pathological processes of the central nervous system, proposing a new theoretical foundation for treating central nervous system diseases.
Collapse
Affiliation(s)
- Haiwen Feng
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin 300070, China
| | - Junjin Li
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin 300070, China
| | - Hongda Wang
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin 300070, China
| | - Zhijian Wei
- Orthopedic Research Center of Shandong University and Department of Orthopedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Shiqing Feng
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin 300070, China
- Orthopedic Research Center of Shandong University and Department of Orthopedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
12
|
Apps JR, Gonzalez-Meljem JM, Guiho R, Pickles JC, Prince E, Schwalbe E, Joshi N, Stone TJ, Ogunbiyi O, Chalker J, Bassey A, Otto G, Davies R, Hughes D, Brandner S, Tan E, Lee V, Hayhurst C, Kline C, Castellano S, Hankinson T, Deutschbein T, Jacques TS, Martinez-Barbera JP. Recurrent adamantinomatous craniopharyngiomas show MAPK pathway activation, clonal evolution and rare TP53-loss-mediated malignant progression. Acta Neuropathol Commun 2024; 12:127. [PMID: 39127699 PMCID: PMC11316312 DOI: 10.1186/s40478-024-01838-4] [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: 06/03/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
The two types of craniopharyngioma, adamantinomatous (ACP) and papillary (PCP), are clinically relevant tumours in children and adults. Although the biology of primary craniopharyngioma is starting to be unravelled, little is known about the biology of recurrence. To fill this gap in knowledge, we have analysed through methylation array, RNA sequencing and pERK1/2 immunohistochemistry a cohort of paired primary and recurrent samples (32 samples from 14 cases of ACP and 4 cases of PCP). We show the presence of copy number alterations and clonal evolution across recurrence in 6 cases of ACP, and analysis of additional whole genome sequencing data from the Children's Brain Tumour Network confirms chromosomal arm copy number changes in at least 7/67 ACP cases. The activation of the MAPK/ERK pathway, a feature previously shown in primary ACP, is observed in all but one recurrent cases of ACP. The only ACP without MAPK activation is an aggressive case of recurrent malignant human craniopharyngioma harbouring a CTNNB1 mutation and loss of TP53. Providing support for a functional role of this TP53 mutation, we show that Trp53 loss in a murine model of ACP results in aggressive tumours and reduced mouse survival. Finally, we characterise the tumour immune infiltrate showing differences in the cellular composition and spatial distribution between ACP and PCP. Together, these analyses have revealed novel insights into recurrent craniopharyngioma and provided preclinical evidence supporting the evaluation of MAPK pathway inhibitors and immunomodulatory approaches in clinical trials in against recurrent ACP.
Collapse
Affiliation(s)
- John R Apps
- Institute of Cancer and Genomic Sciences, Edgbaston Campus, University of Birmingham, Birmingham, B15 2TT, UK.
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK.
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK.
| | - Jose Mario Gonzalez-Meljem
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Tecnologico de Monterrey, School of Engineering and Sciences, Mexico City, Mexico
| | - Romain Guiho
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Oniris, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, Nantes Université, 44000, Nantes, France
| | - Jessica C Pickles
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Eric Prince
- Departments of Neurosurgery and Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, USA
| | - Edward Schwalbe
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Nikhil Joshi
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Thomas J Stone
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Olumide Ogunbiyi
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Jane Chalker
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Akang Bassey
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Georg Otto
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Rosalind Davies
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | | | - Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Enrica Tan
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Victoria Lee
- Sheffield Children's Hospital NHS Foundation Trust, Sheffield, UK
| | | | - Cassie Kline
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sergi Castellano
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Todd Hankinson
- Departments of Neurosurgery and Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, USA
| | - Timo Deutschbein
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
- Medicover Oldenburg MVZ, Oldenburg, Germany
| | - Thomas S Jacques
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
13
|
Chen Y, Liu X, Ainiwan Y, Li M, Pan J, Chen Y, Xiao Z, Wang Z, Xiao X, Tang J, Zeng G, Liang J, Su X, Kungulli R, Fan Y, Lin Q, Liya A, Zheng Y, Chen Z, Xu C, Zhang H, Chen G. Axl as a potential therapeutic target for adamantinomatous craniopharyngiomas: Based on single nucleus RNA-seq and spatial transcriptome profiling. Cancer Lett 2024; 592:216905. [PMID: 38677641 DOI: 10.1016/j.canlet.2024.216905] [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: 12/26/2023] [Revised: 04/07/2024] [Accepted: 04/16/2024] [Indexed: 04/29/2024]
Abstract
Craniopharyngiomas (CPs), particularly Adamantinomatous Craniopharyngiomas (ACPs), often exhibit a heightened risk of postoperative recurrence and severe complications of the endocrine and hypothalamic function. The primary objective of this study is to investigate potential novel targeted therapies within the microenvironment of ACP tumors. Cancer-Associated Fibroblasts (CAFs) were identified in the craniopharyngioma microenvironment, notably in regions characterized by cholesterol clefts, wet keratin, ghost cells, and fibrous stroma in ACPs. CAFs, alongside ghost cells, basaloid-like epithelium cells and calcifications, were found to secrete PROS1 and GAS6, which can activate AXL receptors on the surface of tumor epithelium cells, promoting immune suppression and tumor progression in ACPs. Additionally, the AXL inhibitor Bemcentinib effectively inhibited the proliferation organoids and enhanced the immunotherapeutic efficacy of Atezolizumab. Furthermore, neural crest-like cells were observed in the glial reactive tissue surrounding finger-like protrusions. Overall, our results revealed that the AXL might be a potentially effective therapeutic target for ACPs.
Collapse
Affiliation(s)
- Yiguang Chen
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, 100053, China; Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou North Road, Guangzhou, Guangdong, 510515, China
| | - Xiaohai Liu
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yilamujiang Ainiwan
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou North Road, Guangzhou, Guangdong, 510515, China
| | - Mingchu Li
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jun Pan
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou North Road, Guangzhou, Guangdong, 510515, China
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institute, Stockholm, 10005, Sweden
| | - Zebin Xiao
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Ziyu Wang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
| | - Xinru Xiao
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jie Tang
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Gao Zeng
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jiantao Liang
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xin Su
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Roberta Kungulli
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yuxiang Fan
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Qingtang Lin
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - A Liya
- Guangdong Research Center of Organoid Engineering and Technology, Guangzhou, 510535, China
| | - Yifeng Zheng
- Guangdong Research Center of Organoid Engineering and Technology, Guangzhou, 510535, China
| | - Zexin Chen
- Guangdong Research Center of Organoid Engineering and Technology, Guangzhou, 510535, China
| | - Canli Xu
- Guangdong Research Center of Organoid Engineering and Technology, Guangzhou, 510535, China
| | - Hongqi Zhang
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Ge Chen
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China; China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
14
|
Xu C, Wu J, Ye J, Si Y, Zhang J, Wu B, Pan L, Fu J, Ren Q, Xie S, Tang B, Xiao Y, Hong T. Multiomics integration-based immunological characterizations of adamantinomatous craniopharyngioma in relation to keratinization. Cell Death Dis 2024; 15:439. [PMID: 38906852 PMCID: PMC11192745 DOI: 10.1038/s41419-024-06840-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
Although adamantinomatous craniopharyngioma (ACP) is a tumour with low histological malignancy, there are very few therapeutic options other than surgery. ACP has high histological complexity, and the unique features of the immunological microenvironment within ACP remain elusive. Further elucidation of the tumour microenvironment is particularly important to expand our knowledge of potential therapeutic targets. Here, we performed integrative analysis of 58,081 nuclei through single-nucleus RNA sequencing and spatial transcriptomics on ACP specimens to characterize the features and intercellular network within the microenvironment. The ACP environment is highly immunosuppressive with low levels of T-cell infiltration/cytotoxicity. Moreover, tumour-associated macrophages (TAMs), which originate from distinct sources, highly infiltrate the microenvironment. Using spatial transcriptomic data, we observed one kind of non-microglial derived TAM that highly expressed GPNMB close to the terminally differentiated epithelial cell characterized by RHCG, and this colocalization was verified by asmFISH. We also found the positive correlation of infiltration between these two cell types in datasets with larger cohort. According to intercellular communication analysis, we report a regulatory network that could facilitate the keratinization of RHCG+ epithelial cells, eventually causing tumour progression. Our findings provide a comprehensive analysis of the ACP immune microenvironment and reveal a potential therapeutic strategy base on interfering with these two types of cells.
Collapse
Affiliation(s)
- Chunming Xu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jie Wu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jiye Ye
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yuancheng Si
- Department of Mathematics, University of Manchester, Manchester, UK
- The School of Economics, Fudan University, Shanghai, China
| | - Jinshi Zhang
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Bowen Wu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Laisheng Pan
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jun Fu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Quan Ren
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shenhao Xie
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Bin Tang
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yingqun Xiao
- Department of Pathology, Affiliated Infectious Disease Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Tao Hong
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
15
|
Prince EW, Apps JR, Jeang J, Chee K, Medlin S, Jackson EM, Dudley R, Limbrick D, Naftel R, Johnston J, Feldstein N, Prolo LM, Ginn K, Niazi T, Smith A, Kilburn L, Chern J, Leonard J, Lam S, Hersh DS, Gonzalez-Meljem JM, Amani V, Donson AM, Mitra SS, Bandopadhayay P, Martinez-Barbera JP, Hankinson TC. Unraveling the complexity of the senescence-associated secretory phenotype in adamantinomatous craniopharyngioma using multimodal machine learning analysis. Neuro Oncol 2024; 26:1109-1123. [PMID: 38334125 PMCID: PMC11145462 DOI: 10.1093/neuonc/noae015] [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: 06/13/2023] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Cellular senescence can have positive and negative effects on the body, including aiding in damage repair and facilitating tumor growth. Adamantinomatous craniopharyngioma (ACP), the most common pediatric sellar/suprasellar brain tumor, poses significant treatment challenges. Recent studies suggest that senescent cells in ACP tumors may contribute to tumor growth and invasion by releasing a senesecence-associated secretory phenotype. However, a detailed analysis of these characteristics has yet to be completed. METHODS We analyzed primary tissue samples from ACP patients using single-cell, single-nuclei, and spatial RNA sequencing. We performed various analyses, including gene expression clustering, inferred senescence cells from gene expression, and conducted cytokine signaling inference. We utilized LASSO to select essential gene expression pathways associated with senescence. Finally, we validated our findings through immunostaining. RESULTS We observed significant diversity in gene expression and tissue structure. Key factors such as NFKB, RELA, and SP1 are essential in regulating gene expression, while senescence markers are present throughout the tissue. SPP1 is the most significant cytokine signaling network among ACP cells, while the Wnt signaling pathway predominantly occurs between epithelial and glial cells. Our research has identified links between senescence-associated features and pathways, such as PI3K/Akt/mTOR, MYC, FZD, and Hedgehog, with increased P53 expression associated with senescence in these cells. CONCLUSIONS A complex interplay between cellular senescence, cytokine signaling, and gene expression pathways underlies ACP development. Further research is crucial to understand how these elements interact to create novel therapeutic approaches for patients with ACP.
Collapse
Affiliation(s)
- Eric W Prince
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
- Morgan Adams Foundation for Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - John R Apps
- Oncology Department, Birmingham Women’s and Children’s NHS Foundation Trust, Birmingham, UK
| | - John Jeang
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
- Division of Pediatric Neurosurgery, Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Keanu Chee
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Morgan Adams Foundation for Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Stephen Medlin
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Morgan Adams Foundation for Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Eric M Jackson
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Roy Dudley
- Department of Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - David Limbrick
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robert Naftel
- Department of Neurological Surgery, Vanderbilt University Medical Center, Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, Tennessee, USA
| | - James Johnston
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
- Division of Pediatric Neurosurgery, Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Neil Feldstein
- Department of Neurosurgery, Columbia University Medical Center, New York, New York, USA
| | - Laura M Prolo
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Lucile Packard Children’s Hospital, Stanford University School of Medicine, Palo Alto, California, USA
| | - Kevin Ginn
- The Division of Pediatric Hematology and Oncology, Department of Pediatrics, Children’s Mercy Hospital, Kansas City, Missouri, USA
| | - Toba Niazi
- Department of Pediatric Neurosurgery, Nicklaus Children’s Hospital, Miami, Florida, USA
| | - Amy Smith
- Department of Pediatric Hematology‐Oncology, Arnold Palmer Hospital, Orlando, Florida, USA
| | - Lindsay Kilburn
- Children’s National Health System, Center for Cancer and Blood Disorders, Washington, District of Columbia, USA
- Children’s National Health System, Brain Tumor Institute, Washington, District of Columbia, USA
| | - Joshua Chern
- Departments of Pediatrics and Neurosurgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Pediatric Neurosurgery, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Jeffrey Leonard
- Division of Pediatric Neurosurgery, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Sandi Lam
- Division of Neurosurgery, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | - David S Hersh
- Division of Neurosurgery, Connecticut Children’s, Hartford, Connecticut, USA
| | | | - Vladimir Amani
- Morgan Adams Foundation for Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Andrew M Donson
- Morgan Adams Foundation for Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Siddhartha S Mitra
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Morgan Adams Foundation for Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Pratiti Bandopadhayay
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer, Birth Defects Research Centre, GOS Institute of Child Health, University College London, London, UK
| | - Todd C Hankinson
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
16
|
Lenders NF, Thompson TJ, Chui J, Low J, Inder WJ, Earls PE, McCormack AI. Pituitary tumours without distinct lineage differentiation express stem cell marker SOX2. Pituitary 2024; 27:248-258. [PMID: 38483762 PMCID: PMC11150322 DOI: 10.1007/s11102-024-01385-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/14/2024] [Indexed: 06/05/2024]
Abstract
CONTEXT The recent WHO 2022 Classification of pituitary tumours identified a novel group of 'plurihormonal tumours without distinct lineage differentiation (WDLD)'. By definition, these express multiple combinations of lineage commitment transcription factors, in a monomorphous population of cells. OBJECTIVES To determine the expression of stem cell markers (SOX2, Nestin, CD133) within tumours WDLD, immature PIT-1 lineage and acidophil stem cell tumours, compared with committed cell lineage tumours. METHODS Retrospective evaluation of surgically resected pituitary tumours from St Vincent's Hospital, Sydney. Patients were selected to cover a range of tumour types, based on transcription factor and hormone immunohistochemistry. Clinical data was collected from patient files. Radiology reports were reviewed for size and invasion. Samples were analysed by immunohistochemistry and RT-qPCR for SF-1, PIT-1, T-PIT, SOX2, Nestin and CD133. Stem cell markers were compared between tumours WDLD and those with classically "mature" types. RESULTS On immunohistochemistry, SOX2 was positive in a higher proportion of tumours WDLD compared with those meeting WHO lineage criteria, 7/10 v 10/42 (70 v 23.4%, p = 0.005). CD133 was positive in 2/10 tumours WDLD but 0/41 meeting lineage criteria, P = 0.003. On RT-qPCR, there was no significant difference in relative expression of stem cell markers (SOX2, CD133, Nestin) between tumours with and WDLD. CONCLUSIONS Our study is the first to biologically characterise pituitary tumours WDLD. We demonstrate that these tumours exhibit a higher expression of the stem cell marker SOX2 compared with other lineage-differentiated tumours, suggesting possible involvement of stem cells in their development.
Collapse
Affiliation(s)
- Nèle F Lenders
- Department of Endocrinology, St Vincent's Hospital, Sydney, NSW, Australia.
- Garvan Institute of Medical Research, Level 4, 384 Victoria St, Darlinghurst, NSW, Australia.
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia.
| | - Tanya J Thompson
- Department of Endocrinology, St Vincent's Hospital, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Level 4, 384 Victoria St, Darlinghurst, NSW, Australia
| | - Jeanie Chui
- Department of Anatomical Pathology and Cytopathology, St Vincent's Pathology, Sydney, NSW, Australia
| | - Julia Low
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
- Department of Anatomical Pathology and Cytopathology, St Vincent's Pathology, Sydney, NSW, Australia
| | - Warrick J Inder
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, the University of Queensland, Brisbane, QLD, Australia
| | - Peter E Earls
- Department of Anatomical Pathology and Cytopathology, St Vincent's Pathology, Sydney, NSW, Australia
| | - Ann I McCormack
- Department of Endocrinology, St Vincent's Hospital, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Level 4, 384 Victoria St, Darlinghurst, NSW, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
17
|
Jiang B, Zhang W, Zhang X, Sun Y. Targeting senescent cells to reshape the tumor microenvironment and improve anticancer efficacy. Semin Cancer Biol 2024; 101:58-73. [PMID: 38810814 DOI: 10.1016/j.semcancer.2024.05.002] [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: 01/17/2024] [Revised: 05/09/2024] [Accepted: 05/12/2024] [Indexed: 05/31/2024]
Abstract
Cancer is daunting pathology with remarkable breadth and scope, spanning genetics, epigenetics, proteomics, metalobomics and cell biology. Cellular senescence represents a stress-induced and essentially irreversible cell fate associated with aging and various age-related diseases, including malignancies. Senescent cells are characterized of morphologic alterations and metabolic reprogramming, and develop a highly active secretome termed as the senescence-associated secretory phenotype (SASP). Since the first discovery, senescence has been understood as an important barrier to tumor progression, as its induction in pre-neoplastic cells limits carcinogenesis. Paradoxically, senescent cells arising in the tumor microenvironment (TME) contribute to tumor progression, including augmented therapeutic resistance. In this article, we define typical forms of senescent cells commonly observed within the TME and how senescent cells functionally remodel their surrounding niche, affect immune responses and promote cancer evolution. Furthermore, we highlight the recently emerging pipelines of senotherapies particularly senolytics, which can selectively deplete senescent cells from affected organs in vivo and impede tumor progression by restoring therapeutic responses and securing anticancer efficacies. Together, co-targeting cancer cells and their normal but senescent counterparts in the TME holds the potential to achieve increased therapeutic benefits and restrained disease relapse in future clinical oncology.
Collapse
Affiliation(s)
- Birong Jiang
- School of Pharmacy, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Wei Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xuguang Zhang
- Mengniu Institute of Nutrition Science, Global R&D Innovation Center, Shanghai 200124, China
| | - Yu Sun
- School of Pharmacy, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong 264003, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China; Department of Medicine and VAPSHCS, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
18
|
Wang X, Zhao C, Lin J, Liu H, Zeng Q, Chen H, Wang Y, Xu D, Chen W, Xu M, Zhang E, Lin D, Lin Z. Multi-omics analysis of adamantinomatous craniopharyngiomas reveals distinct molecular subgroups with prognostic and treatment response significance. Chin Med J (Engl) 2024; 137:859-870. [PMID: 37565822 PMCID: PMC10997223 DOI: 10.1097/cm9.0000000000002774] [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: 05/30/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND Adamantinomatous craniopharyngioma (ACP) is the commonest pediatric sellar tumor. No effective drug is available and interpatient heterogeneity is prominent. This study aimed to identify distinct molecular subgroups of ACP based on the multi-omics profiles, imaging findings, and histological features, in order to predict the response to anti-inflammatory treatment and immunotherapies. METHODS Totally 142 Chinese cases diagnosed with craniopharyngiomas were profiled, including 119 ACPs and 23 papillary craniopharyngiomas. Whole-exome sequencing (151 tumors, including recurrent ones), RNA sequencing (84 tumors), and DNA methylome profiling (95 tumors) were performed. Consensus clustering and non-negative matrix factorization were used for subgrouping, and Cox regression were utilized for prognostic evaluation, respectively. RESULTS Three distinct molecular subgroups were identified: WNT, ImA, and ImB. The WNT subgroup showed higher Wnt/β-catenin pathway activity, with a greater number of epithelial cells and more predominantly solid tumors. The ImA and ImB subgroups had activated inflammatory and interferon response pathways, with enhanced immune cell infiltration and more predominantly cystic tumors. Mitogen-activated protein kinases (MEK/MAPK) signaling was activated only in ImA samples, while IL-6 and epithelial-mesenchymal transition biomarkers were highly expressed in the ImB group, mostly consisting of children. The degree of astrogliosis was significantly elevated in the ImA group, with severe finger-like protrusions at the invasive front of the tumor. The molecular subgrouping was an independent prognostic factor, with the WNT group having longer event-free survival than ImB (Cox, P = 0.04). ImA/ImB cases were more likely to respond to immune checkpoint blockade (ICB) therapy than the WNT group ( P <0.01). In the preliminary screening of subtyping markers, CD38 was significantly downregulated in WNT compared with ImA and ImB ( P = 0.01). CONCLUSIONS ACP comprises three molecular subtypes with distinct imaging and histological features. The prognosis of the WNT type is better than that of the ImB group, which is more likely to benefit from the ICB treatment.
Collapse
Affiliation(s)
- Xianlong Wang
- Department of Bioinformatics, School of Medical Technology and Engineering, Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Chuan Zhao
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Jincheng Lin
- Department of Bioinformatics, School of Medical Technology and Engineering, Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Hongxing Liu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Qiuhong Zeng
- Department of Bioinformatics, School of Medical Technology and Engineering, Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Huadong Chen
- Department of Bioinformatics, School of Medical Technology and Engineering, Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Ye Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Dapeng Xu
- Department of Bioinformatics, School of Medical Technology and Engineering, Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Wen Chen
- Department of Bioinformatics, School of Medical Technology and Engineering, Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Moping Xu
- Department of Bioinformatics, School of Medical Technology and Engineering, Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - En Zhang
- Department of Bioinformatics, School of Medical Technology and Engineering, Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Da Lin
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Zhixiong Lin
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| |
Collapse
|
19
|
Fan G, Yu B, Tang L, Zhu R, Chen J, Zhu Y, Huang H, Zhou L, Liu J, Wang W, Tao Z, Zhang F, Yu S, Lu X, Cao Y, Du S, Li H, Li J, Zhang J, Ren H, Gires O, Liu H, Wang X, Qin J, Wang H. TSPAN8 + myofibroblastic cancer-associated fibroblasts promote chemoresistance in patients with breast cancer. Sci Transl Med 2024; 16:eadj5705. [PMID: 38569015 DOI: 10.1126/scitranslmed.adj5705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 03/06/2024] [Indexed: 04/05/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are abundant stromal cells in the tumor microenvironment that promote cancer progression and relapse. However, the heterogeneity and regulatory roles of CAFs underlying chemoresistance remain largely unclear. Here, we performed a single-cell analysis using high-dimensional flow cytometry analysis and identified a distinct senescence-like tetraspanin-8 (TSPAN8)+ myofibroblastic CAF (myCAF) subset, which is correlated with therapeutic resistance and poor survival in multiple cohorts of patients with breast cancer (BC). TSPAN8+ myCAFs potentiate the stemness of the surrounding BC cells through secretion of senescence-associated secretory phenotype (SASP)-related factors IL-6 and IL-8 to counteract chemotherapy. NAD-dependent protein deacetylase sirtuin 6 (SIRT6) reduction was responsible for the senescence-like phenotype and tumor-promoting role of TSPAN8+ myCAFs. Mechanistically, TSPAN8 promoted the phosphorylation of ubiquitin E3 ligase retinoblastoma binding protein 6 (RBBP6) at Ser772 by recruiting MAPK11, thereby inducing SIRT6 protein destruction. In turn, SIRT6 down-regulation up-regulated GLS1 and PYCR1, which caused TSPAN8+ myCAFs to secrete aspartate and proline, and therefore proved a nutritional niche to support BC outgrowth. By demonstrating that TSPAN8+SIRT6low myCAFs were tightly associated with unfavorable disease outcomes, we proposed that the combined regimen of anti-TSPAN8 antibody and SIRT6 activator MDL-800 is a promising approach to overcome chemoresistance. These findings highlight that senescence contributes to CAF heterogeneity and chemoresistance and suggest that targeting TSPAN8+ myCAFs is a promising approach to circumvent chemoresistance.
Collapse
Affiliation(s)
- Guangjian Fan
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Bo Yu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lei Tang
- Department of Oncology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou 215000, China
| | - Rongxuan Zhu
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Jianhua Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ying Zhu
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - He Huang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200243, China
| | - Liying Zhou
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200243, China
| | - Jun Liu
- Department of Breast-thyroid Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Wei Wang
- Department of Breast-thyroid Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zhonghua Tao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fengchun Zhang
- Department of Oncology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou 215000, China
| | - Siwei Yu
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xiaoqing Lu
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China
| | - Yuan Cao
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Shaoqian Du
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Huihui Li
- Department of Breast Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province 271016, China
| | - Junjian Li
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Jian Zhang
- Key Laboratory of Cell Differentiation and Apoptosis, Ministry of Education, Department of Pathophysiology, Ruijin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai 342500, China
| | - He Ren
- Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Olivier Gires
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, LMU, Munich 80336, Germany
| | - Haikun Liu
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Xin Wang
- Department of Surgery, Chinese University of Hong Kong Prince of Wales Hospital, Shatin, Hong Kong SAR 999077, China
| | - Jun Qin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Hongxia Wang
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
20
|
Sun R, Feng J, Wang J. Underlying Mechanisms and Treatment of Cellular Senescence-Induced Biological Barrier Interruption and Related Diseases. Aging Dis 2024; 15:612-639. [PMID: 37450933 PMCID: PMC10917536 DOI: 10.14336/ad.2023.0621] [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: 04/03/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
Given its increasing prevalence, aging is of great concern to researchers worldwide. Cellular senescence is a physiological or pathological cellular state caused by aging and a prominent risk factor for the interruption of the integrity and functionality of human biological barriers. Health barriers play an important role in maintaining microenvironmental homeostasis within the body. The senescence of barrier cells leads to barrier dysfunction and age-related diseases. Cellular senescence has been reported to be a key target for the prevention of age-related barrier diseases, including Alzheimer's disease, Parkinson's disease, age-related macular degeneration, diabetic retinopathy, and preeclampsia. Drugs such as metformin, dasatinib, quercetin, BCL-2 inhibitors, and rapamycin have been shown to intervene in cellular senescence and age-related diseases. In this review, we conclude that cellular senescence is involved in age-related biological barrier impairment. We further outline the cellular pathways and mechanisms underlying barrier impairment caused by cellular senescence and describe age-related barrier diseases associated with senescent cells. Finally, we summarize the currently used anti-senescence pharmacological interventions and discuss their therapeutic potential for preventing age-related barrier diseases.
Collapse
Affiliation(s)
- Ruize Sun
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, China
| | - Jue Wang
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
21
|
Chen Z, Zhou J, Wu Y, Chen F, Li J, Tao L, Tian Y, Wang H, Li J, Li Z, He W, Zhang K, Wang H. METTL3 promotes cellular senescence of colorectal cancer via modulation of CDKN2B transcription and mRNA stability. Oncogene 2024; 43:976-991. [PMID: 38361047 DOI: 10.1038/s41388-024-02956-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 01/19/2024] [Accepted: 01/25/2024] [Indexed: 02/17/2024]
Abstract
Cellular senescence plays a critical role in cancer development, but the underlying mechanisms remain poorly understood. Our recent study uncovered that replicative senescent colorectal cancer (CRC) cells exhibit increased levels of mRNA N6-methyladenosine (m6A) and methyltransferase METTL3. Knockdown of METTL3 can restore the senescence-associated secretory phenotype (SASP) of CRC cells. Our findings, which were confirmed by m6A-sequencing and functional studies, demonstrate that the cyclin-dependent kinase inhibitor 2B (CDKN2B, encoding p15INK4B) is a mediator of METTL3-regulated CRC senescence. Specifically, m6A modification at position A413 in the coding sequence (CDS) of CDKN2B positively regulates its mRNA stability by recruiting IGF2BP3 and preventing binding with the CCR4-NOT complex. Moreover, increased METTL3 methylates and stabilizes the mRNA of E2F1, which binds to the -208 to -198 regions of the CDKN2B promoter to facilitate transcription. Inhibition of METTL3 or specifically targeting CDKN2B methylation can suppress CRC senescence. Finally, the METTL3/CDKN2B axis-induced senescence can facilitate M2 macrophage polarization and is correlated with aging and CRC progression. The involvement of METTL3/CDKN2B in cell senescence provides a new potential therapeutic target for CRC treatment and expands our understanding of mRNA methylation's role in cellular senescence.
Collapse
Affiliation(s)
- Zhuojia Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Jiawang Zhou
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - You Wu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Feng Chen
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jianing Li
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Lijun Tao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yifan Tian
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Haoran Wang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jiexin Li
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zigang Li
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518067, China
| | - Weiling He
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China.
| | - Kun Zhang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu Seventh People's Hospital, Affiliated Cancer Hospital of Chengdu Medical College, School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China.
| | - Hongsheng Wang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
22
|
Pérez Millán MI, Cheung LYM, Mercogliano F, Camilletti MA, Chirino Felker GT, Moro LN, Miriuka S, Brinkmeier ML, Camper SA. Pituitary stem cells: past, present and future perspectives. Nat Rev Endocrinol 2024; 20:77-92. [PMID: 38102391 PMCID: PMC10964491 DOI: 10.1038/s41574-023-00922-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/31/2023] [Indexed: 12/17/2023]
Abstract
Pituitary cells that express the transcription factor SOX2 are stem cells because they can self-renew and differentiate into multiple pituitary hormone-producing cell types as organoids. Wounding and physiological challenges can activate pituitary stem cells, but cell numbers are not fully restored, and the ability to mobilize stem cells decreases with increasing age. The basis of these limitations is still unknown. The regulation of stem cell quiescence and activation involves many different signalling pathways, including those mediated by WNT, Hippo and several cytokines; more research is needed to understand the interactions between these pathways. Pituitary organoids can be formed from human or mouse embryonic stem cells, or from human induced pluripotent stem cells. Human pituitary organoid transplantation is sufficient to induce corticosterone release in hypophysectomized mice, raising the possibility of therapeutic applications. Today, pituitary organoids have the potential to assess the role of individual genes and genetic variants on hormone production ex vivo, providing an important tool for the advancement of exciting frontiers in pituitary stem cell biology and pituitary organogenesis. In this article, we provide an overview of notable discoveries in pituitary stem cell function and highlight important areas for future research.
Collapse
Affiliation(s)
- María Inés Pérez Millán
- Institute of Bioscience, Biotechnology and Translational Biology (IB3-UBA), University of Buenos Aires, Buenos Aires, Argentina
| | - Leonard Y M Cheung
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Physiology and Biophysics, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Florencia Mercogliano
- Institute of Bioscience, Biotechnology and Translational Biology (IB3-UBA), University of Buenos Aires, Buenos Aires, Argentina
| | - Maria Andrea Camilletti
- Institute of Bioscience, Biotechnology and Translational Biology (IB3-UBA), University of Buenos Aires, Buenos Aires, Argentina
| | - Gonzalo T Chirino Felker
- Laboratory of Applied Research of Neurosciences (LIAN-CONICET), FLENI Sede Escobar, Buenos Aires, Argentina
| | - Lucia N Moro
- Laboratory of Applied Research of Neurosciences (LIAN-CONICET), FLENI Sede Escobar, Buenos Aires, Argentina
| | - Santiago Miriuka
- Laboratory of Applied Research of Neurosciences (LIAN-CONICET), FLENI Sede Escobar, Buenos Aires, Argentina
| | - Michelle L Brinkmeier
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Sally A Camper
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
23
|
Gonzalez-Meljem JM, Martinez-Barbera JP. Implications of cellular senescence in paediatric pituitary tumours. EBioMedicine 2024; 99:104905. [PMID: 38043401 PMCID: PMC10730348 DOI: 10.1016/j.ebiom.2023.104905] [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: 08/03/2023] [Revised: 11/20/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023] Open
Abstract
The long-standing view of senescent cells as passive and dysfunctional biological remnants has recently shifted into a new paradigm where they are main players in the development of many diseases, including cancer. The senescence programme represents a first line of defence that prevents tumour cell growth but also leads to the secretion of multiple pro-inflammatory and pro-tumourigenic factors that fuel tumour initiation, growth, and progression. Here, we review the main molecular features and biological functions of senescent cells in cancer, including the outcomes of inducing or targeting senescence. We discuss evidence on the role of cellular senescence in pituitary tumours, with an emphasis on adamantinomatous craniopharyngioma (ACP) and pituitary adenomas. Although senescence has been proposed to be a tumour-preventing mechanism in pituitary adenomas, research in ACP has shown that senescent cells are tumour-promoting in both murine models and human tumours. Future studies characterizing the impact of targeting senescent cells may result in novel therapies against pituitary tumours.
Collapse
Affiliation(s)
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Institute of Child Health, London, UK.
| |
Collapse
|
24
|
McHugh D, Sun B, Gutierrez-Muñoz C, Hernández-González F, Mellone M, Guiho R, Duran I, Pombo J, Pietrocola F, Birch J, Kallemeijn WW, Khadayate S, Dharmalingam G, Vernia S, Tate EW, Martínez-Barbera JP, Withers DJ, Thomas GJ, Serrano M, Gil J. COPI vesicle formation and N-myristoylation are targetable vulnerabilities of senescent cells. Nat Cell Biol 2023; 25:1804-1820. [PMID: 38012402 PMCID: PMC10709147 DOI: 10.1038/s41556-023-01287-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 10/12/2023] [Indexed: 11/29/2023]
Abstract
Drugs that selectively kill senescent cells (senolytics) improve the outcomes of cancer, fibrosis and age-related diseases. Despite their potential, our knowledge of the molecular pathways that affect the survival of senescent cells is limited. To discover senolytic targets, we performed RNAi screens and identified coatomer complex I (COPI) vesicle formation as a liability of senescent cells. Genetic or pharmacological inhibition of COPI results in Golgi dispersal, dysfunctional autophagy, and unfolded protein response-dependent apoptosis of senescent cells, and knockdown of COPI subunits improves the outcomes of cancer and fibrosis in mouse models. Drugs targeting COPI have poor pharmacological properties, but we find that N-myristoyltransferase inhibitors (NMTi) phenocopy COPI inhibition and are potent senolytics. NMTi selectively eliminated senescent cells and improved outcomes in models of cancer and non-alcoholic steatohepatitis. Our results suggest that senescent cells rely on a hyperactive secretory apparatus and that inhibiting trafficking kills senescent cells with the potential to treat various senescence-associated diseases.
Collapse
Affiliation(s)
- Domhnall McHugh
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Bin Sun
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Carmen Gutierrez-Muñoz
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Fernanda Hernández-González
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Department of Pulmonology, ICR, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Massimiliano Mellone
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- AstraZeneca, Immuno-Oncology Discovery, Oncology R&D, Cambridge, UK
| | - Romain Guiho
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Imanol Duran
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Joaquim Pombo
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Federico Pietrocola
- Karolinska Institute, Department of Biosciences and Nutrition, Huddinge, Sweden
| | - Jodie Birch
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Wouter W Kallemeijn
- Department of Chemistry, Molecular Sciences Research Hub, London, UK
- The Francis Crick Institute, London, UK
| | - Sanjay Khadayate
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Gopuraja Dharmalingam
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Santiago Vernia
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Edward W Tate
- Department of Chemistry, Molecular Sciences Research Hub, London, UK
- The Francis Crick Institute, London, UK
| | - Juan Pedro Martínez-Barbera
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Dominic J Withers
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Gareth J Thomas
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Altos Labs, Cambridge Institute of Science, Granta Park, UK
| | - Jesús Gil
- MRC Laboratory of Medical Sciences (LMS), London, UK.
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
25
|
Jin L, Cai K, Wu W, Xiao Y, Qiao N, Liu F, Ru S, Cao L, Zhu H, Bai J, Liu C, Li C, Zhao P, Zhang Y, Gui S. Correlations between the expression of molecules in the TGF-β signaling pathway and clinical factors in adamantinomatous craniopharyngiomas. Front Endocrinol (Lausanne) 2023; 14:1167776. [PMID: 37854185 PMCID: PMC10579895 DOI: 10.3389/fendo.2023.1167776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 08/31/2023] [Indexed: 10/20/2023] Open
Abstract
Objective To investigate the clinical and pathological factors associated with preoperative hypothalamus invasion and postoperative outcomes of adamantinomatous craniopharyngiomas (ACPs) after the expanded endonasal approach (EEA) resection. Methods Ninety-three specimens of ACPs, consisting of 71 primary and 22 recurrent tumors, were investigated for the expression of TGF-β1, SMAD2, SMAD3, and β-catenin by immunohistochemistry staining. The clinical information of relevant patients, including the extent of resection, hypothalamus invasion, endocrinopathy, complications, and prognosis, was reviewed. The relationships between the expression of these immunopathological markers and clinical factors were analyzed. Results Endocrinological dysfunctions were more common in recurrent patients and primary patients with hypothalamus invasion in the comparisons. For recurrent patients, the rate of gross total resection (GTR) was significantly lower than for primary patients (63.6% vs. 90.1%, P = 0.007). According to radiological and intraoperative findings, invasive ACPs (IACPs) included 48 (67.6%) cases in primary tumors. The expression of TGF-β1 and β-catenin was significantly higher in recurrent tumors (P = 0.021 and P = 0.018, respectively) and IACPs (P = 0.008 and P = 0.004, respectively). The expression level of TGF-β1 was associated with hypothalamus involvement (Puget grade, P = 0.05; Vile grade, P = 0.002), postoperative endocrinopathy (P = 0.01), and pituitary stalk preservation (P = 0.008) in primary patients. In addition, the extent of resection, treatment history, hypothalamic invasion, and level of TGF-β1 expression had significant influences on tumor recurrence/progression after surgery separately. Conclusion Our study demonstrated the potential role of TGF-β1 in the regulation of hypothalamus invasion in ACPs and the prediction of prognosis after EEA surgery. The TGF-β signaling pathway may represent a crucial mechanism in the aggressive behavior and progression of ACPs.
Collapse
Affiliation(s)
- Lu Jin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kefan Cai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - WenTao Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Youchao Xiao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ning Qiao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fangzheng Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Siming Ru
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lei Cao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Haibo Zhu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiwei Bai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chunhui Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Peng Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Laporte E, Vankelecom H. Organoid models of the pituitary gland in health and disease. Front Endocrinol (Lausanne) 2023; 14:1233714. [PMID: 37614709 PMCID: PMC10442803 DOI: 10.3389/fendo.2023.1233714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/20/2023] [Indexed: 08/25/2023] Open
Abstract
The pituitary gland represents the hub of our endocrine system. Its cells produce specific hormones that direct multiple vital physiological processes such as body growth, fertility, and stress. The gland also contains a population of stem cells which are still enigmatic in phenotype and function. Appropriate research models are needed to advance our knowledge on pituitary (stem cell) biology. Over the last decade, 3D organoid models have been established, either derived from the pituitary stem cells or from pluripotent stem cells, covering both healthy and diseased conditions. Here, we summarize the state-of-the-art of pituitary-allied organoid models and discuss applications of these powerful in vitro research and translational tools to study pituitary development, biology, and disease.
Collapse
Affiliation(s)
- Emma Laporte
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Laboratory of Tissue Plasticity in Health and Disease, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Hugo Vankelecom
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Laboratory of Tissue Plasticity in Health and Disease, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| |
Collapse
|
27
|
D'Ambrosio M, Gil J. Reshaping of the tumor microenvironment by cellular senescence: An opportunity for senotherapies. Dev Cell 2023; 58:1007-1021. [PMID: 37339603 DOI: 10.1016/j.devcel.2023.05.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 02/13/2023] [Accepted: 05/19/2023] [Indexed: 06/22/2023]
Abstract
Cellular senescence is a stress response associated with aging and disease, including cancer. Senescent cells undergo a stable cell cycle arrest, undergo a change in morphology and metabolic reprogramming, and produce a bioactive secretome termed the senescence-associated secretory phenotype (SASP). In cancer, senescence is an important barrier to tumor progression. Induction of senescence in preneoplastic cells limits cancer initiation, and many cancer therapies act in part by inducing senescence in cancer cells. Paradoxically, senescent cells lingering in the tumor microenvironment (TME) can contribute to tumor progression, metastasis, and therapy resistance. In this review, we discuss the different types of senescent cells present in the TME and how these senescent cells and their SASP reshape the TME, affect immune responses, and influence cancer progression. Furthermore, we will highlight the importance of senotherapies, including senolytic drugs that eliminate senescent cells and impede tumor progression and metastasis by restoring anti-tumor immune responses and influencing the TME.
Collapse
Affiliation(s)
- Mariantonietta D'Ambrosio
- MRC London Institute of Medical Sciences (LMS), Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Jesús Gil
- MRC London Institute of Medical Sciences (LMS), Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
28
|
Han YP, Lin HW, Li H. Cancer Stem Cells in Tumours of the Central Nervous System in Children: A Comprehensive Review. Cancers (Basel) 2023; 15:3154. [PMID: 37370764 DOI: 10.3390/cancers15123154] [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: 04/14/2023] [Revised: 05/30/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer stem cells (CSCs) are a subgroup of cells found in various kinds of tumours with stem cell characteristics, such as self-renewal, induced differentiation, and tumourigenicity. The existence of CSCs is regarded as a major source of tumour recurrence, metastasis, and resistance to conventional chemotherapy and radiation treatment. Tumours of the central nervous system (CNS) are the most common solid tumours in children, which have many different types including highly malignant embryonal tumours and midline gliomas, and low-grade gliomas with favourable prognoses. Stem cells from the CNS tumours have been largely found and reported by researchers in the last decade and their roles in tumour biology have been deeply studied. However, the cross-talk of CSCs among different CNS tumour types and their clinical impacts have been rarely discussed. This article comprehensively reviews the achievements in research on CSCs in paediatric CNS tumours. Biological functions, diagnostic values, and therapeutic perspectives are reviewed in detail. Further investigations into CSCs are warranted to improve the clinical practice in treating children with CNS tumours.
Collapse
Affiliation(s)
- Yi-Peng Han
- Department of Neurosurgery, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Hou-Wei Lin
- Department of Paediatric Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Department of Paediatric Surgery, Jiaxing Women and Children Hospital Affiliated to Jiaxing University, Jiaxing 314001, China
| | - Hao Li
- Department of Neurosurgery, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| |
Collapse
|
29
|
Apps JR, Muller HL, Hankinson TC, Yock TI, Martinez-Barbera JP. Contemporary Biological Insights and Clinical Management of Craniopharyngioma. Endocr Rev 2023; 44:518-538. [PMID: 36574377 DOI: 10.1210/endrev/bnac035] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 11/20/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Craniopharyngiomas (CPs) are clinically aggressive tumors because of their invasive behavior and recalcitrant tendency to recur after therapy. There are 2 types based on their distinct histology and molecular features: the papillary craniopharyngioma (PCP), which is associated with BRAF-V600E mutations and the adamantinomatous craniopharyngioma (ACP), characterized by mutations in CTNNB1 (encoding β-catenin). Patients with craniopharyngioma show symptoms linked to the location of the tumor close to the optic pathways, hypothalamus, and pituitary gland, such as increased intracranial pressure, endocrine deficiencies, and visual defects. Treatment is not specific and mostly noncurative, and frequently includes surgery, which may achieve gross total or partial resection, followed by radiotherapy. In cystic tumors, frequent drainage is often required and intracystic instillation of drugs has been used to help manage cyst refilling. More recently targeted therapies have been used, particularly in PCP, but also now in ACP and clinical trials are underway or in development. Although patient survival is high, the consequences of the tumor and its treatment can lead to severe comorbidities resulting in poor quality of life, in particular for those patients who bear tumors with hypothalamic involvement. Accordingly, in these patients at risk for the development of a hypothalamic syndrome, hypothalamus-sparing treatment strategies such as limited resection followed by irradiation are recommended. In this review, we provide an update on various aspects of CP, with emphasis on recent advances in the understanding of tumor pathogenesis, clinical consequences, management, and therapies.
Collapse
Affiliation(s)
- John Richard Apps
- Institute of Cancer and Genomics Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- Developmental Biology and Cancer, Birth Defects Research Centre, GOS Institute of Child Health, University College London, London, WC1N 1EH, UK
- Oncology Department, Birmingham Women's and Children's NHS Foundation Trust, Birmingham B4 6NH, UK
| | - Hermann Lothar Muller
- Department of Pediatrics and Pediatric Hematology/Oncology, University Children's Hospital, Carl von Ossietzky University, Klinikum Oldenburg AöR, 26133 Oldenburg, Germany
| | - Todd Cameron Hankinson
- Department of Neurosurgery, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- Department of Pediatric Neurosurgery, Children's Hospital Colorado, University of Colorado, Aurora, Colorado 80045, USA
- Morgan Adams Foundation Pediatric Brain Tumor Program, Aurora, Colorado, USA
| | - Torunn Ingrid Yock
- Department of Radiation Oncology, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02115, USA
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer, Birth Defects Research Centre, GOS Institute of Child Health, University College London, London, WC1N 1EH, UK
| |
Collapse
|
30
|
Jiang Y, Yang J, Liang R, Zan X, Fan R, Shan B, Liu H, Li L, Wang Y, Wu M, Qi X, Chen H, Ren Q, Liu Z, Wang Y, Zhang J, Zhou P, Li Q, Tian M, Yang J, Wang C, Li X, Jiang S, Zhou L, Zhang G, Chen Y, Xu J. Single-cell RNA sequencing highlights intratumor heterogeneity and intercellular network featured in adamantinomatous craniopharyngioma. SCIENCE ADVANCES 2023; 9:eadc8933. [PMID: 37043580 PMCID: PMC10096597 DOI: 10.1126/sciadv.adc8933] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 03/10/2023] [Indexed: 06/19/2023]
Abstract
Despite improvements in microscopically neurosurgical techniques made in recent years, the prognosis of adamantinomatous craniopharyngioma (ACP) is still unsatisfactory. Little is known about cellular atlas and biological features of ACP. Here, we carried out integrative analysis of 44,038 single-cell transcriptome profiles to characterize the landscape of intratumoral heterogeneity and tumor microenvironment (TME) in ACP. Four major neoplastic cell states with distinctive expression signatures were defined, which further revealed the histopathological features and elucidated unknown cellular atlas of ACP. Pseudotime analyses suggested potential evolutionary trajectories between specific neoplastic cell states. Notably, a distinct oligodendrocyte lineage was identified in ACP, which was associated with immunological infiltration and neural damage. In addition, we described a tumor-centric regulatory network based on intercellular communication in TME. Together, our findings represent a unique resource for deciphering tumor heterogeneity of ACP, which will improve clinical diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinlong Yang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ruichao Liang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Zan
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rangrang Fan
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Baoyin Shan
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Li
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yue Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, 250000, China
| | - Min Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Qi
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hongxu Chen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qingqing Ren
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiyong Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuelong Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Peizhi Zhou
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiang Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Tian
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinhao Yang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chaoyang Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xueying Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shu Jiang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liangxue Zhou
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gao Zhang
- Faculty of Dentistry, The University of Hong Kong, Sai Ying Pun, 999077, Hong Kong
| | - Yaohui Chen
- Department of Thoracic Surgery/Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
31
|
Wang C, Zhang H, Fan J, Li Q, Guo R, Pan J, Liu Y, Peng J, Zhu Q, Feng Y, Wu C, Luo P, Qiu X, Shi J, Deng Y, Qi S, Liu Y. Inhibition of integrated stress response protects against lipid-induced senescence in hypothalamic neural stem cells in adamantinomatous craniopharyngioma. Neuro Oncol 2023; 25:720-732. [PMID: 36454228 PMCID: PMC10076952 DOI: 10.1093/neuonc/noac261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Adamantinomatous craniopharyngioma (ACP) is a benign tumor with malignant clinical manifestations. ACP adjacent to the hypothalamus often presents with more severe symptoms and higher incidence of hypothalamic dysfunction. However, the mechanism underlying hypothalamic dysfunction remains unclear. METHODS Immunostaining was performed to determine the nerve damage to the floor of the third ventricle (3VF) adjacent to ACP and to examine the recruitment and senescence of hypothalamic neural stem cells (htNSCs). The accumulation of lipid droplets (LDs) in htNSCs was evaluated via BODIPY staining, oil red O staining, and transmission electron microscopy. In vitro and in vivo assays were used to evaluate the effect of cystic fluid or oxidized low-density lipoprotein and that of oxytocin (OXT) on htNSC senescence and the hypothalamic function. The protein expression levels were analyzed using western blotting. RESULTS htNSCs with massive LD accumulation were recruited to the damaged 3VF adjacent to ACP. The LDs in htNSCs induced senescence and reduced neuronal differentiation; however, htNSC senescence was effectively prevented by inhibiting either CD36 or integrated stress response (ISR) signaling. Furthermore, OXT pretreatment reduced lipotoxicity via the inhibition of ISR signaling and the repair of the blood-brain barrier. CONCLUSIONS Reduced LD aggregation or ISR signaling inhibition prevented senescence in htNSCs and identified molecular pathways and potential therapeutic targets that may improve hypothalamic dysfunction in ACP patients.
Collapse
Affiliation(s)
- Chaohu Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Huarong Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Jun Fan
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Qing Li
- Department of Dietetics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Rongrong Guo
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Jun Pan
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yawei Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Junxiang Peng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Qianchao Zhu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yiwen Feng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Chengdong Wu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Peng Luo
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xiaoyu Qiu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Jin Shi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yingying Deng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yi Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
- Central Laboratory, Shunde Hospital, Southern Medical University, Foshan 528300, Guangdong, China
| |
Collapse
|
32
|
Li Z, Tian M, Wang G, Cui X, Ma J, Liu S, Shen B, Liu F, Wu K, Xiao X, Zhu C. Senotherapeutics: An emerging approach to the treatment of viral infectious diseases in the elderly. Front Cell Infect Microbiol 2023; 13:1098712. [PMID: 37065192 PMCID: PMC10094634 DOI: 10.3389/fcimb.2023.1098712] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/06/2023] [Indexed: 03/31/2023] Open
Abstract
In the context of the global COVID-19 pandemic, the phenomenon that the elderly have higher morbidity and mortality is of great concern. Existing evidence suggests that senescence and viral infection interact with each other. Viral infection can lead to the aggravation of senescence through multiple pathways, while virus-induced senescence combined with existing senescence in the elderly aggravates the severity of viral infections and promotes excessive age-related inflammation and multiple organ damage or dysfunction, ultimately resulting in higher mortality. The underlying mechanisms may involve mitochondrial dysfunction, abnormal activation of the cGAS-STING pathway and NLRP3 inflammasome, the role of pre-activated macrophages and over-recruited immune cells, and accumulation of immune cells with trained immunity. Thus, senescence-targeted drugs were shown to have positive effects on the treatment of viral infectious diseases in the elderly, which has received great attention and extensive research. Therefore, this review focused on the relationship between senescence and viral infection, as well as the significance of senotherapeutics for the treatment of viral infectious diseases.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mingfu Tian
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Guolei Wang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xianghua Cui
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun’e Ma
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Siyu Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Bingzheng Shen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fang Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xuan Xiao
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Chengliang Zhu, ; Xuan Xiao,
| | - Chengliang Zhu
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Chengliang Zhu, ; Xuan Xiao,
| |
Collapse
|
33
|
Zhao B, Wu B, Feng N, Zhang X, Zhang X, Wei Y, Zhang W. Aging microenvironment and antitumor immunity for geriatric oncology: the landscape and future implications. J Hematol Oncol 2023; 16:28. [PMID: 36945046 PMCID: PMC10032017 DOI: 10.1186/s13045-023-01426-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/15/2023] [Indexed: 03/23/2023] Open
Abstract
The tumor microenvironment (TME) has been extensively investigated; however, it is complex and remains unclear, especially in elderly patients. Senescence is a cellular response to a variety of stress signals, which is characterized by stable arrest of the cell cycle and major changes in cell morphology and physiology. To the best of our knowledge, senescence leads to consistent arrest of tumor cells and remodeling of the tumor-immune microenvironment (TIME) by activating a set of pleiotropic cytokines, chemokines, growth factors, and proteinases, which constitute the senescence-associated secretory phenotype (SASP). On the one hand, the SASP promotes antitumor immunity, which enhances treatment efficacy; on the other hand, the SASP increases immunosuppressive cell infiltration, including myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs), M2 macrophages, and N2 neutrophils, contributing to TIME suppression. Therefore, a deeper understanding of the regulation of the SASP and components contributing to robust antitumor immunity in elderly individuals with different cancer types and the available therapies is necessary to control tumor cell senescence and provide greater clinical benefits to patients. In this review, we summarize the key biological functions mediated by cytokines and intercellular interactions and significant components of the TME landscape, which influence the immunotherapy response in geriatric oncology. Furthermore, we summarize recent advances in clinical practices targeting TME components and discuss potential senescent TME targets.
Collapse
Affiliation(s)
- Binghao Zhao
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Departments of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100032, China
| | - Bo Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Nan Feng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiang Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xin Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yiping Wei
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, China.
| |
Collapse
|
34
|
Campanini ML, Almeida JP, Martins CS, de Castro M. The molecular pathogenesis of craniopharyngiomas. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2023; 67:266-275. [PMID: 36748936 PMCID: PMC10689043 DOI: 10.20945/2359-3997000000600] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/11/2022] [Indexed: 02/08/2023]
Abstract
Research from the last 20 years has provided important insights into the molecular pathogenesis of craniopharyngiomas (CPs). Besides the well-known clinical and histological differences between the subtypes of CPs, adamantinomatous (ACP) and papillary (PCP) craniopharyngiomas, other molecular differences have been identified, further elucidating pathways related to the origin and development of such tumors. The present minireview assesses current knowledge on embryogenesis and the genetic, epigenetic, transcriptomic, and signaling pathways involved in the ACP and PCP subtypes, revealing the similarities and differences in their profiles. ACP and PCP subtypes can be identified by the presence of mutations in CTNNB1 and BRAF genes, with prevalence around 60% and 90%, respectively. Therefore, β-catenin accumulates in the nucleus-cytoplasm of cell clusters in ACPs and, in PCPs, cell immunostaining with specific antibody against the V600E-mutated protein can be seen. Distinct patterns of DNA methylation further differentiate ACPs and PCPs. In addition, research on genetic and epigenetic changes and tumor microenvironment specificities have further clarified the development and progression of the disease. No relevant transcriptional differences in ACPs have emerged between children and adults. In conclusion, ACPs and PCPs present diverse genetic signatures and each subtype is associated with specific signaling pathways. A better understanding of the pathways related to the growth of such tumors is paramount for the development of novel targeted therapeutic agents.
Collapse
Affiliation(s)
- Marina Lanciotti Campanini
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil,
| | - João Paulo Almeida
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, United States
| | - Clarissa Silva Martins
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
- Faculdade de Medicina, Universidade Federal do Mato Grosso do Sul, Campo Grande, RS, Brasil
| | - Margaret de Castro
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| |
Collapse
|
35
|
Bou-Nassif R, El Ahmadieh TY, Abou-Mrad Z, Rosenblum M, Cohen MA, Tabar V. De novo adamantinomatous craniopharyngioma with long-term pre-diagnostic imaging. Neurochirurgie 2023; 69:101404. [PMID: 36701980 DOI: 10.1016/j.neuchi.2023.101404] [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: 10/04/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/26/2023]
Abstract
Adamantinomatous craniopharyngioma has a bimodal age distribution occurring in children aged 5-15 years and less frequently in adults aged 45-60 years. The current embryogenetic hypothesis suggests that adamantinomatous craniopharyngioma (ACP) arises from epithelial remnants of the craniopharyngeal duct or Rathke's pouch. It is thought that this tumor exists early on during childhood but remains indolent, growing very slowly until it is diagnosed incidentally or due to symptoms. Recent reports of de novo development of ACP, however, have challenged this theory. Herein, we present a case of an incidentally discovered de novo adamantinomatous craniopharyngioma that was documented to arise de novo on serial MRIs performed for a different indication. To our knowledge, this is the first report of a middle-aged patient who is diagnosed with a de novo ACP documented with contrast-enhanced MRIs of the sella over a 16-year period. This case challenges our current understanding of the pathophysiology of adamantinomatous craniopharyngioma.
Collapse
Affiliation(s)
- R Bou-Nassif
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, USA; Multidisciplinary Pituitary and Skull Base Tumor Program, Memorial Sloan Kettering Cancer Center, New York, USA.
| | - T Y El Ahmadieh
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, USA; Multidisciplinary Pituitary and Skull Base Tumor Program, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Z Abou-Mrad
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, USA; Multidisciplinary Pituitary and Skull Base Tumor Program, Memorial Sloan Kettering Cancer Center, New York, USA
| | - M Rosenblum
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - M A Cohen
- Multidisciplinary Pituitary and Skull Base Tumor Program, Memorial Sloan Kettering Cancer Center, New York, USA; Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - V Tabar
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, USA; Multidisciplinary Pituitary and Skull Base Tumor Program, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
36
|
López-Otín C, Pietrocola F, Roiz-Valle D, Galluzzi L, Kroemer G. Meta-hallmarks of aging and cancer. Cell Metab 2023; 35:12-35. [PMID: 36599298 DOI: 10.1016/j.cmet.2022.11.001] [Citation(s) in RCA: 230] [Impact Index Per Article: 115.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/11/2022] [Accepted: 11/07/2022] [Indexed: 01/05/2023]
Abstract
Both aging and cancer are characterized by a series of partially overlapping "hallmarks" that we subject here to a meta-analysis. Several hallmarks of aging (i.e., genomic instability, epigenetic alterations, chronic inflammation, and dysbiosis) are very similar to specific cancer hallmarks and hence constitute common "meta-hallmarks," while other features of aging (i.e., telomere attrition and stem cell exhaustion) act likely to suppress oncogenesis and hence can be viewed as preponderantly "antagonistic hallmarks." Disabled macroautophagy and cellular senescence are two hallmarks of aging that exert context-dependent oncosuppressive and pro-tumorigenic effects. Similarly, the equivalence or antagonism between aging-associated deregulated nutrient-sensing and cancer-relevant alterations of cellular metabolism is complex. The agonistic and antagonistic relationship between the processes that drive aging and cancer has bearings for the age-related increase and oldest age-related decrease of cancer morbidity and mortality, as well as for the therapeutic management of malignant disease in the elderly.
Collapse
Affiliation(s)
- Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| | - Federico Pietrocola
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - David Roiz-Valle
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
37
|
Domen A, Deben C, Verswyvel J, Flieswasser T, Prenen H, Peeters M, Lardon F, Wouters A. Cellular senescence in cancer: clinical detection and prognostic implications. J Exp Clin Cancer Res 2022; 41:360. [PMID: 36575462 PMCID: PMC9793681 DOI: 10.1186/s13046-022-02555-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/30/2022] [Indexed: 12/28/2022] Open
Abstract
Cellular senescence is a state of stable cell-cycle arrest with secretory features in response to cellular stress. Historically, it has been considered as an endogenous evolutionary homeostatic mechanism to eliminate damaged cells, including damaged cells which are at risk of malignant transformation, thereby protecting against cancer. However, accumulation of senescent cells can cause long-term detrimental effects, mainly through the senescence-associated secretory phenotype, and paradoxically contribute to age-related diseases including cancer. Besides its role as tumor suppressor, cellular senescence is increasingly being recognized as an in vivo response in cancer patients to various anticancer therapies. Its role in cancer is ambiguous and even controversial, and senescence has recently been promoted as an emerging hallmark of cancer because of its hallmark-promoting capabilities. In addition, the prognostic implications of cellular senescence have been underappreciated due to the challenging detection and sparse in and ex vivo evidence of cellular senescence in cancer patients, which is only now catching up. In this review, we highlight the approaches and current challenges of in and ex vivo detection of cellular senescence in cancer patients, and we discuss the prognostic implications of cellular senescence based on in and ex vivo evidence in cancer patients.
Collapse
Affiliation(s)
- Andreas Domen
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610, Wilrijk (Antwerp), Belgium.
- Department of Oncology, Antwerp University Hospital (UZA), 2650, Edegem (Antwerp), Belgium.
| | - Christophe Deben
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610, Wilrijk (Antwerp), Belgium
| | - Jasper Verswyvel
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610, Wilrijk (Antwerp), Belgium
| | - Tal Flieswasser
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610, Wilrijk (Antwerp), Belgium
| | - Hans Prenen
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610, Wilrijk (Antwerp), Belgium
- Department of Oncology, Antwerp University Hospital (UZA), 2650, Edegem (Antwerp), Belgium
| | - Marc Peeters
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610, Wilrijk (Antwerp), Belgium
- Department of Oncology, Antwerp University Hospital (UZA), 2650, Edegem (Antwerp), Belgium
| | - Filip Lardon
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610, Wilrijk (Antwerp), Belgium
| | - An Wouters
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610, Wilrijk (Antwerp), Belgium
| |
Collapse
|
38
|
Aminzadeh-Gohari S, Kofler B, Herzog C. Dietary restriction in senolysis and prevention and treatment of disease. Crit Rev Food Sci Nutr 2022; 64:5242-5268. [PMID: 36484738 PMCID: PMC7616065 DOI: 10.1080/10408398.2022.2153355] [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] [Indexed: 12/13/2022]
Abstract
Aging represents a key risk factor for a plethora of diseases. Targeting detrimental processes which occur during aging, especially before onset of age-related disease, could provide drastic improvements in healthspan. There is increasing evidence that dietary restriction (DR), including caloric restriction, fasting, or fasting-mimicking diets, extend both lifespan and healthspan. This has sparked interest in the use of dietary regimens as a non-pharmacological means to slow aging and prevent disease. Here, we review the current evidence on the molecular mechanisms underlying DR-induced health improvements, including removal of senescent cells, metabolic reprogramming, and epigenetic rejuvenation.
Collapse
Affiliation(s)
- Sepideh Aminzadeh-Gohari
- Research Program for Receptor Biochemistry and Tumor Metabollism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Research Institute for Biomedical Ageing, Universität Innsbruck, Innsbruck, Austria
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabollism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Chiara Herzog
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Research Institute for Biomedical Ageing, Universität Innsbruck, Innsbruck, Austria
| |
Collapse
|
39
|
Saleh T, Khasawneh AI, Himsawi N, Abu-Raideh J, Ejeilat V, Elshazly AM, Gewirtz DA. Senolytic Therapy: A Potential Approach for the Elimination of Oncogene-Induced Senescent HPV-Positive Cells. Int J Mol Sci 2022; 23:15512. [PMID: 36555154 PMCID: PMC9778669 DOI: 10.3390/ijms232415512] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Senescence represents a unique cellular stress response characterized by a stable growth arrest, macromolecular alterations, and wide spectrum changes in gene expression. Classically, senescence is the end-product of progressive telomeric attrition resulting from the repetitive division of somatic cells. In addition, senescent cells accumulate in premalignant lesions, in part, as a product of oncogene hyperactivation, reflecting one element of the tumor suppressive function of senescence. Oncogenic processes that induce senescence include overexpression/hyperactivation of H-Ras, B-Raf, and cyclin E as well as inactivation of PTEN. Oncogenic viruses, such as Human Papilloma Virus (HPV), have also been shown to induce senescence. High-risk strains of HPV drive the immortalization, and hence transformation, of cervical epithelial cells via several mechanisms, but primarily via deregulation of the cell cycle, and possibly, by facilitating escape from senescence. Despite the wide and successful utilization of HPV vaccines in reducing the incidence of cervical cancer, this measure is not effective in preventing cancer development in individuals already positive for HPV. Accordingly, in this commentary, we focus on the potential contribution of oncogene and HPV-induced senescence (OIS) in cervical cancer. We further consider the potential utility of senolytic agents for the elimination of HPV-harboring senescent cells as a strategy for reducing HPV-driven transformation and the risk of cervical cancer development.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
| | - Ashraf I. Khasawneh
- Department of Microbiology, Pathology, and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
| | - Nisreen Himsawi
- Department of Microbiology, Pathology, and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
| | - Jumana Abu-Raideh
- Department of Microbiology, Pathology, and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
| | - Vera Ejeilat
- Department of Anatomy and Histology, Faculty of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Ahmed M. Elshazly
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
40
|
Willis TL, Lodge EJ, Andoniadou CL, Yianni V. Cellular interactions in the pituitary stem cell niche. Cell Mol Life Sci 2022; 79:612. [PMID: 36451046 PMCID: PMC9712314 DOI: 10.1007/s00018-022-04612-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 09/27/2022] [Accepted: 10/25/2022] [Indexed: 12/03/2022]
Abstract
Stem cells in the anterior pituitary gland can give rise to all resident endocrine cells and are integral components for the appropriate development and subsequent maintenance of the organ. Located in discreet niches within the gland, stem cells are involved in bi-directional signalling with their surrounding neighbours, interactions which underpin pituitary gland homeostasis and response to organ challenge or physiological demand. In this review we highlight core signalling pathways that steer pituitary progenitors towards specific endocrine fate decisions throughout development. We further elaborate on those which are conserved in the stem cell niche postnatally, including WNT, YAP/TAZ and Notch signalling. Furthermore, we have collated a directory of single cell RNA sequencing studies carried out on pituitaries across multiple organisms, which have the potential to provide a vast database to study stem cell niche components in an unbiased manner. Reviewing published data, we highlight that stem cells are one of the main signalling hubs within the anterior pituitary. In future, coupling single cell sequencing approaches with genetic manipulation tools in vivo, will enable elucidation of how previously understudied signalling pathways function within the anterior pituitary stem cell niche.
Collapse
Affiliation(s)
- Thea L Willis
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Emily J Lodge
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Cynthia L Andoniadou
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK.
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| | - Val Yianni
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK.
| |
Collapse
|
41
|
Bousset L, Gil J. Targeting senescence as an anticancer therapy. Mol Oncol 2022; 16:3855-3880. [PMID: 36065138 PMCID: PMC9627790 DOI: 10.1002/1878-0261.13312] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/12/2022] [Accepted: 08/21/2022] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a stress response elicited by different molecular insults. Senescence results in cell cycle exit and is characterised by multiple phenotypic changes such as the production of a bioactive secretome. Senescent cells accumulate during ageing and are present in cancerous and fibrotic lesions. Drugs that selectively kill senescent cells (senolytics) have shown great promise for the treatment of age-related diseases. Senescence plays paradoxical roles in cancer. Induction of senescence limits cancer progression and contributes to therapy success, but lingering senescent cells fuel progression, recurrence, and metastasis. In this review, we describe the intricate relation between senescence and cancer. Moreover, we enumerate how current anticancer therapies induce senescence in tumour cells and how senolytic agents could be deployed to complement anticancer therapies. "One-two punch" therapies aim to first induce senescence in the tumour followed by senolytic treatment to target newly exposed vulnerabilities in senescent tumour cells. "One-two punch" represents an emerging and promising new strategy in cancer treatment. Future challenges of "one-two punch" approaches include how to best monitor senescence in cancer patients to effectively survey their efficacy.
Collapse
Affiliation(s)
- Laura Bousset
- MRC London Institute of Medical Sciences (LMS)UK
- Faculty of Medicine, Institute of Clinical Sciences (ICS)Imperial College LondonUK
| | - Jesús Gil
- MRC London Institute of Medical Sciences (LMS)UK
- Faculty of Medicine, Institute of Clinical Sciences (ICS)Imperial College LondonUK
| |
Collapse
|
42
|
Abstract
Senescence is a cellular response to a variety of stress signals that is characterized by a stable withdrawal from the cell cycle and major changes in cell morphology and physiology. While most research on senescence has been performed on non-cancer cells, it is evident that cancer cells can also mount a senescence response. In this Review, we discuss how senescence can be induced in cancer cells. We describe the distinctive features of senescent cancer cells and how these changes in cellular physiology might be exploited for the selective eradication of these cells (senolysis). We discuss activation of the host immune system as a particularly attractive way to clear senescent cancer cells. Finally, we consider the challenges and opportunities provided by a 'one-two punch' sequential treatment of cancer with pro-senescence therapy followed by senolytic therapy.
Collapse
Affiliation(s)
- Liqin Wang
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lina Lankhorst
- Cancer, Stem Cells & Developmental Biology programme, Utrecht University, Utrecht, The Netherlands
| | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Müller HL, Tauber M, Lawson EA, Özyurt J, Bison B, Martinez-Barbera JP, Puget S, Merchant TE, van Santen HM. Hypothalamic syndrome. Nat Rev Dis Primers 2022; 8:24. [PMID: 35449162 DOI: 10.1038/s41572-022-00351-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/08/2022] [Indexed: 12/11/2022]
Abstract
Hypothalamic syndrome (HS) is a rare disorder caused by disease-related and/or treatment-related injury to the hypothalamus, most commonly associated with rare, non-cancerous parasellar masses, such as craniopharyngiomas, germ cell tumours, gliomas, cysts of Rathke's pouch and Langerhans cell histiocytosis, as well as with genetic neurodevelopmental syndromes, such as Prader-Willi syndrome and septo-optic dysplasia. HS is characterized by intractable weight gain associated with severe morbid obesity, multiple endocrine abnormalities and memory impairment, attention deficit and reduced impulse control as well as increased risk of cardiovascular and metabolic disorders. Currently, there is no cure for this condition but treatments for general obesity are often used in patients with HS, including surgery, medication and counselling. However, these are mostly ineffective and no medications that are specifically approved for the treatment of HS are available. Specific challenges in HS are because the syndrome represents an adverse effect of different diseases, and that diagnostic criteria, aetiology, pathogenesis and management of HS are not completely defined.
Collapse
Affiliation(s)
- Hermann L Müller
- Department of Paediatrics and Paediatric Hematology/Oncology, University Children's Hospital, Klinikum Oldenburg AöR, Carl von Ossietzky University, Oldenburg, Germany.
| | - Maithé Tauber
- Centre de Référence du Syndrome de Prader-Willi et autres syndromes avec troubles du comportement alimentaire, Hôpital des Enfants, CHU-Toulouse, Toulouse, France
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity) INSERM UMR1291 - CNRS UMR5051 - Université Toulouse III, Toulouse, France
| | - Elizabeth A Lawson
- Neuroendocrine Unit, Massachusetts General Hospital, and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jale Özyurt
- Biological Psychology Laboratory, Department of Psychology, School of Medicine and Health Sciences, Carl von Ossietzky University, Oldenburg, Germany
- Research Center Neurosensory Science, Carl von Ossietzky University, Oldenburg, Germany
| | - Brigitte Bison
- Department of Neuroradiology, University Hospital Augsburg, Augsburg, Germany
| | - Juan-Pedro Martinez-Barbera
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Stephanie Puget
- Service de Neurochirurgie, Hôpital Necker-Enfants Malades, Sorbonne Paris Cité, Paris, France
- Service de Neurochirurgie, Hopital Pierre Zobda Quitman, Martinique, France
| | - Thomas E Merchant
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hanneke M van Santen
- Department of Paediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center, Utrecht, Netherlands
- Princess Máxima Center for Paediatric Oncology, Utrecht, Netherlands
| |
Collapse
|
44
|
Senotherapeutics in Cancer and HIV. Cells 2022; 11:cells11071222. [PMID: 35406785 PMCID: PMC8997781 DOI: 10.3390/cells11071222] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 12/14/2022] Open
Abstract
Cellular senescence is a stress-response mechanism that contributes to homeostasis maintenance, playing a beneficial role during embryogenesis and in normal adult organisms. In contrast, chronic senescence activation may be responsible for other events such as age-related disorders, HIV and cancer development. Cellular senescence activation can be triggered by different insults. Regardless of the inducer, there are several phenotypes generally shared among senescent cells: cell division arrest, an aberrant shape, increased size, high granularity because of increased numbers of lysosomes and vacuoles, apoptosis resistance, defective metabolism and some chromatin alterations. Senescent cells constitute an important area for research due to their contributions to the pathogenesis of different diseases such as frailty, sarcopenia and aging-related diseases, including cancer and HIV infection, which show an accelerated aging. Hence, a new pharmacological category of treatments called senotherapeutics is under development. This group includes senolytic drugs that selectively attack senescent cells and senostatic drugs that suppress SASP factor delivery, inhibiting senescent cell development. These new drugs can have positive therapeutic effects on aging-related disorders and act in cancer as antitumor drugs, avoiding the undesired effects of senescent cells such as those from SASP. Here, we review senotherapeutics and how they might affect cancer and HIV disease, two very different aging-related diseases, and review some compounds acting as senolytics in clinical trials.
Collapse
|
45
|
Mikuła-Pietrasik J, Rutecki S, Książek K. The functional multipotency of transforming growth factor β signaling at the intersection of senescence and cancer. Cell Mol Life Sci 2022; 79:196. [PMID: 35305149 PMCID: PMC11073081 DOI: 10.1007/s00018-022-04236-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/22/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022]
Abstract
The transforming growth factor β (TGF-β) family of cytokines comprises a group of proteins, their receptors, and effector molecules that, in a coordinated manner, modulate a plethora of physiological and pathophysiological processes. TGF-β1 is the best known and plausibly most active representative of this group. It acts as an immunosuppressant, contributes to extracellular matrix remodeling, and stimulates tissue fibrosis, differentiation, angiogenesis, and epithelial-mesenchymal transition. In recent years, this cytokine has been established as a vital regulator of organismal aging and cellular senescence. Finally, the role of TGF-β1 in cancer progression is no longer in question. Because this protein is involved in so many, often overlapping phenomena, the question arises whether it can be considered a molecular bridge linking some of these phenomena together and governing their reciprocal interactions. In this study, we reviewed the literature from the perspective of the role of various TGF-β family members as regulators of a complex mutual interplay between senescence and cancer. These aspects are then considered in a broader context of remaining TGF-β-related functions and coexisting processes. The main narrative axis in this work is centered around the interaction between the senescence of normal peritoneal cells and ovarian cancer cells. The discussion also includes examples of TGF-β activity at the interface of other normal and cancer cell types.
Collapse
Affiliation(s)
- Justyna Mikuła-Pietrasik
- Department of Pathophysiology of Ageing and Civilization Diseases, Długa ½ Str, Poznań University of Medical Sciences, 61-848, Poznań, Poland
| | - Szymon Rutecki
- Department of Pathophysiology of Ageing and Civilization Diseases, Długa ½ Str, Poznań University of Medical Sciences, 61-848, Poznań, Poland
| | - Krzysztof Książek
- Department of Pathophysiology of Ageing and Civilization Diseases, Długa ½ Str, Poznań University of Medical Sciences, 61-848, Poznań, Poland.
| |
Collapse
|
46
|
Morsli S, Doherty GJ, Muñoz-Espín D. Activatable senoprobes and senolytics: Novel strategies to detect and target senescent cells. Mech Ageing Dev 2022; 202:111618. [PMID: 34990647 DOI: 10.1016/j.mad.2021.111618] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 01/10/2023]
Abstract
Pharmacologically active compounds that manipulate cellular senescence (senotherapies) have recently shown great promise in multiple pre-clinical disease models, and some of them are now being tested in clinical trials. Despite promising proof-of-principle evidence, there are known on- and off-target toxicities associated with these compounds, and therefore more refined and novel strategies to improve their efficacy and specificity for senescent cells are being developed. Preferential release of drugs and macromolecular formulations within senescent cells has been predominantly achieved by exploiting one of the most widely used biomarkers of senescence, the increase in lysosomal senescence-associated β-galactosidase (SA-β-gal) activity, a common feature of most reported senescent cell types. Galacto-conjugation is a versatile therapeutic and detection strategy to facilitate preferential targeting of senescent cells by using a variety of existing formulations, including modular systems, nanocarriers, activatable prodrugs, probes, and small molecules. We discuss the benefits and drawbacks of these specific senescence targeting tools and how the strategy of galacto-conjugation might be utilised to design more specific and sophisticated next-generation senotherapeutics, as well as theranostic agents. Finally, we discuss some innovative strategies and possible future directions for the field.
Collapse
Affiliation(s)
- Samir Morsli
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK
| | - Gary J Doherty
- Department of Oncology, Box 193, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK.
| | - Daniel Muñoz-Espín
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK.
| |
Collapse
|
47
|
Alexandraki KI, Xekouki P. Medical Therapy for Craniopharyngiomas. TOUCHREVIEWS IN ENDOCRINOLOGY 2022; 17:121-132. [PMID: 35118458 DOI: 10.17925/ee.2021.17.2.121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/01/2021] [Indexed: 11/24/2022]
Abstract
Craniopharyngiomas are rare benign neoplasms presenting in two different types, adamantinomatous (ACP) or papillary (PCP), which are molecularly and clinically distinct. Traditional treatment includes surgical resection and radiotherapy, which are accompanied by a number of debilitating complications because of the tumours' proximity to important brain structures. Recent advances in the understanding of molecular pathogenesis of craniopharyngiomas have opened horizons to medical therapeutic options. ACPs are mainly characterized by mutations of β-catenin, which activate Wingless/Int (Wnt), and alter the mitogen extracellular kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway, as well as inflammatory, cellular senescence, programmed cell death and sonic hedgehog (SHH) pathways. PCPs are mainly characterized by Ras/Raf/MEK/ERK pathway activation secondary to BRAF-V600E mutations. MEK inhibitors, such as binimetinib, or anti-inflammatory mediators, such as tocilizumab or interferon, have been administered to patients with ACP and the efficacy is mostly favourable. On the other hand, BRAF inhibitors, such as dabrafenib or vemurafenib, either alone or in combination with the MEK inhibitors trametinib and cobimetinib, have been administered to patients with PCP resulting in favourable responses. A number of ongoing trials will shed light on schemes, doses, combined treatments and safety issues of the new molecular-targeted treatments, changing the management of patients with craniopharyngiomas by launching the era of personalized medicine in these rare neoplasms. We conducted a systematic review to identify case series or case reports with patients currently treated with systemic medical therapy.
Collapse
Affiliation(s)
- Krystallenia I Alexandraki
- Second Department of Surgery, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Paraskevi Xekouki
- Endocrinology and Diabetes Clinic, University General Hospital of Heraklion, University of Crete School of Medicine, Heraklion, Greece
| |
Collapse
|
48
|
Zhao R, Jin X, Li A, Xu B, Shen Y, Wang W, Huang J, Zhang Y, Li X. Precise Diabetic Wound Therapy: PLS Nanospheres Eliminate Senescent Cells via DPP4 Targeting and PARP1 Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104128. [PMID: 34738744 PMCID: PMC8728814 DOI: 10.1002/advs.202104128] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Indexed: 05/14/2023]
Abstract
Diabetic ulcers, a difficult problem faced by clinicians, are strongly associated with an increase in cellular senescence. Few empirical studies have focused on exploring a targeted strategy to cure diabetic wounds by eliminating senescent fibroblasts (SFs) and reducing side effects. In this study, poly-l-lysine/sodium alginate (PLS) is modified with talabostat (PT100) and encapsulates a PARP1 plasmid (PARP1@PLS-PT100) for delivery to target the dipeptidyl peptidase 4 (DPP4) receptor and eliminate SFs. PARP1@PLS-PT100 releases encapsulated plasmids, displaying high selectivity for SFs over normal fibroblasts by targeting the DPP4 receptor, decreasing senescence-associated secretory phenotypes (SASPs), and stimulating the secretion of anti-inflammatory factors. Furthermore, the increased apoptosis of SFs and the disappearance of cellular senescence alleviates SASPs, accelerates re-epithelialization and collagen deposition, and significantly induces macrophage M2 polarization, which mediates tissue repair and the inflammatory response. This innovative strategy has revealed the previously undefined role of PARP1@PLS-PT100 in promoting diabetic wound healing, suggesting its therapeutic potential in refractory wound repair.
Collapse
Affiliation(s)
- Renliang Zhao
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Xiangyun Jin
- Department of Orthopedic TraumaRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127P. R. China
| | - Ang Li
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Bitong Xu
- Department of SpineCenter for Orthopaedic SurgeryThe Third Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Yifan Shen
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Wei Wang
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Jinghuan Huang
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yadong Zhang
- Department of SpineCenter for Orthopaedic SurgeryThe Third Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Xiaolin Li
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| |
Collapse
|
49
|
Hu MC, Moe OW. Phosphate and Cellular Senescence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1362:55-72. [PMID: 35288873 PMCID: PMC10513121 DOI: 10.1007/978-3-030-91623-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cellular senescence is one type of permeant arrest of cell growth and one of increasingly recognized contributor to aging and age-associated disease. High phosphate and low Klotho individually and synergistically lead to age-related degeneration in multiple organs. Substantial evidence supports the causality of high phosphate in cellular senescence, and potential contribution to human aging, cancer, cardiovascular, kidney, neurodegenerative, and musculoskeletal diseases. Phosphate can induce cellular senescence both by direct phosphotoxicity, and indirectly through downregulation of Klotho and upregulation of plasminogen activator inhibitor-1. Restriction of dietary phosphate intake and blockage of intestinal absorption of phosphate help suppress cellular senescence. Supplementation of Klotho protein, cellular senescence inhibitor, and removal of senescent cells with senolytic agents are potential novel strategies to attenuate phosphate-induced cellular senescence, retard aging, and ameliorate age-associated, and phosphate-induced disorders.
Collapse
Affiliation(s)
- Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Departments of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
50
|
Saleh T, Carpenter VJ. Potential Use of Senolytics for Pharmacological Targeting of Precancerous Lesions. Mol Pharmacol 2021; 100:580-587. [PMID: 34544896 DOI: 10.1124/molpharm.121.000361] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/10/2021] [Indexed: 01/10/2023] Open
Abstract
Senescence is a cell state that contributes to several homeostatic and pathologic processes. In addition to being induced in somatic cells in response to replicative exhaustion (replicative senescence) as part of organismal aging, senescence can also be triggered prematurely by oncogene hyperactivation or tumor suppressor dysfunction [oncogene-induced senescence (OIS)]. Consequently, senescent cells comprise a major component of precancerous lesions of skin, oral mucosa, nasopharynx, prostate, gut, and lung. Unfortunately, invasive (or minimally invasive) interventions are currently the only available approach employed to eradicate premalignant lesions that carry the potential for cancer progression. Senolytics are a newly emerging drug class capable of selectively eliminating senescent cells. Although senolytics have been successfully demonstrated to mitigate a myriad of aging-related pathologies and to cull senescent cancer cells, there is a paucity of evidence for the potential use of senolytics as a novel approach to eliminate oncogene-induced senescent cells. This Emerging Concepts commentary will 1) summarize evidence in established models of OIS including B-Raf-induced nevi, transgenic lung cancer, and pancreatic adenocarcinoma models, as well as evidence from clinical precancerous lesions; 2) suggest that OIS is targetable; and 3) propose the utilization of senolytic agents as a revolutionary means to interfere with the ability of senescent premalignant cells to progress to cancer in vitro and in vivo If proven to be effective, senolytics will represent an emerging tool to pharmacologically treat precancerous lesions. SIGNIFICANCE STATEMENT: The treatment of premalignant lesions is largely based on the utilization of invasive (or minimally invasive) measures. Oncogene-induced senescence (OIS) is one form of senescence that occurs in response to oncogene overexpression in somatic cells and is present in precancerous lesions. Although the contribution of OIS to disease progression is undetermined, recent evidence suggests that senescent cells are permissive for malignant transformation. Accordingly, the pharmacological targeting of oncogene-induced senescent cells could potentially provide a novel, less invasive, means for the treatment of premalignant disease.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, Hashemite University, Zarqa, Jordan (T.S.); Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia (V.J.C.)
| | - Valerie J Carpenter
- Department of Basic Medical Sciences, Faculty of Medicine, Hashemite University, Zarqa, Jordan (T.S.); Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia (V.J.C.)
| |
Collapse
|