Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31.802
Filter
1.
Front Immunol ; 15: 1382931, 2024.
Article in English | MEDLINE | ID: mdl-38736882

ABSTRACT

Background: Neuroblastoma (NB) is characterized by both adrenergic (ADRN) and undifferentiated mesenchymal (MES) subsets. The ganglioside sialic acid-containing glycosphingolipid (GD2) is widely overexpressed on tumors of neuroectodermal origin promoting malignant phenotypes. MES cells are greatly enriched in post-therapy and relapsing tumors and are characterized by decreased expression of GD2. This event may cause failure of GD2-based immunotherapy. NK cells represent a key innate cell subset able to efficiently kill tumors. However, the tumor microenvironment (TME) that includes tumor cells and tumor-associated (TA) cells could inhibit their effector function. Methods: We studied eight NB primary cultures that, in comparison with commercial cell lines, more faithfully reflect the tumor cell characteristics. We studied four primary NB-MES cell cultures and two pairs of MES/ADRN (691 and 717) primary cultures, derived from the same patient. In particular, in the six human NB primary cultures, we assessed their phenotype, the expression of GD2, and the enzymes that control its expression, as well as their interactions with NK cells, using flow cytometry, RT-qPCR, and cytotoxicity assays. Results: We identified mature (CD105+/CD133-) and undifferentiated (CD133+/CD105-) NB subsets that express high levels of the MES transcripts WWTR1 and SIX4. In addition, undifferentiated MES cells display a strong resistance to NK-mediated killing. On the contrary, mature NB-MES cells display an intermediate resistance to NK-mediated killing and exhibit some immunomodulatory capacities on NK cells but do not inhibit their cytolytic activity. Notably, independent from their undifferentiated or mature phenotype, NB-MES cells express GD2 that can be further upregulated in undifferentiated NB-MES cells upon co-culture with NK cells, leading to the generation of mature mesenchymal GD2bright neuroblasts. Concerning 691 and 717, they show high levels of GD2 and resistance to NK cell-mediated killing that can be overcome by the administration of dinutuximab beta, the anti-GD2 monoclonal antibody applied in the clinic. Conclusions: NB is a heterogeneous tumor representing a further hurdle in NB immunotherapy. However, different from what was reported with NB commercial cells and independent of their MES/ADRN phenotype, the expression of GD2 and its displayed sensitivity to anti-GD2 mAb ADCC indicated the possible effectiveness of anti-GD2 immunotherapy.


Subject(s)
Gangliosides , Killer Cells, Natural , Neuroblastoma , Tumor Escape , Tumor Microenvironment , Humans , Neuroblastoma/immunology , Neuroblastoma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Gangliosides/immunology , Gangliosides/metabolism , Tumor Microenvironment/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Tumor Cells, Cultured , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism
2.
HLA ; 103(5): e15515, 2024 May.
Article in English | MEDLINE | ID: mdl-38747019

ABSTRACT

Although a number of susceptibility loci for neuroblastoma (NB) have been identified by genome-wide association studies, it is still unclear whether variants in the HLA region contribute to NB susceptibility. In this study, we conducted a comprehensive genetic analysis of variants in the HLA region among 724 NB patients and 2863 matched controls from different cohorts. We exploited whole-exome sequencing data to accurately type HLA alleles with an ensemble approach on the results from three different typing tools, and carried out rigorous sample quality control to ensure a fine-scale ancestry matching. The frequencies of common HLA alleles were compared between cases and controls by logistic regression under additive and non-additive models. Population stratification was taken into account adjusting for ancestry-informative principal components. We detected significant HLA associations with NB. In particular, HLA-DQB1*05:02 (OR = 1.61; padj = 5.4 × 10-3) and HLA-DRB1*16:01 (OR = 1.60; padj = 2.3 × 10-2) alleles were associated to higher risk of developing NB. Conditional analysis highlighted the HLA-DQB1*05:02 allele and its residue Ser57 as key to this association. DQB1*05:02 allele was not associated to clinical features worse outcomes in the NB cohort. Nevertheless, a risk score derived from the allelic combinations of five HLA variants showed a substantial predictive value for patient survival (HR = 1.53; p = 0.032) that was independent from established NB prognostic factors. Our study leveraged powerful computational methods to explore WES data and HLA variants and to reveal complex genetic associations. Further studies are needed to validate the mechanisms of these interactions that contribute to the multifaceted pattern of factors underlying the disease initiation and progression.


Subject(s)
Alleles , Exome Sequencing , Genetic Predisposition to Disease , Neuroblastoma , Humans , Neuroblastoma/genetics , Neuroblastoma/mortality , Exome Sequencing/methods , Case-Control Studies , Male , Female , Gene Frequency , HLA-DQ beta-Chains/genetics , HLA Antigens/genetics , Genome-Wide Association Study , HLA-DRB1 Chains/genetics , Polymorphism, Single Nucleotide
3.
Cancer Immunol Immunother ; 73(7): 122, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714539

ABSTRACT

Neuroblastoma (NB) is the most common and deadliest extracranial solid tumor in children. Targeting tumor-associated macrophages (TAMs) is a strategy for attenuating tumor-promoting states. The crosstalk between cancer cells and TAMs plays a pivotal role in mediating tumor progression in NB. The overexpression of Hexokinase-3 (HK3), a pivotal enzyme in glucose metabolism, has been associated with poor prognosis in NB patients. Furthermore, it correlates with the infiltration of M2-like macrophages within NB tumors, indicating its significant involvement in tumor progression. Therefore, HK3 not only directly regulates the malignant biological behaviors of tumor cells, such as proliferation, migration, and invasion, but also recruits and polarizes M2-like macrophages through the PI3K/AKT-CXCL14 axis in neuroblastoma. The secretion of lactate and histone lactylation alterations within tumor cells accompanies this interaction. Additionally, elevated expression of HK3 in M2-TAMs was found at the same time. Modulating HK3 within M2-TAMs alters the biological behavior of tumor cells, as demonstrated by our in vitro studies. This study highlights the pivotal role of HK3 in the progression of NB malignancy and its intricate regulatory network with M2-TAMs. It establishes HK3 as a promising dual-functional biomarker and therapeutic target in combating neuroblastoma.


Subject(s)
Hexokinase , Neuroblastoma , Tumor-Associated Macrophages , Neuroblastoma/metabolism , Neuroblastoma/pathology , Humans , Hexokinase/metabolism , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Cell Proliferation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Phosphatidylinositol 3-Kinases/metabolism , Cell Line, Tumor , Cell Movement , Chemokines, CXC/metabolism , Animals , Tumor Microenvironment/immunology
4.
Clin Transl Med ; 14(5): e1680, 2024 May.
Article in English | MEDLINE | ID: mdl-38769668

ABSTRACT

BACKGROUND: A series of studies have demonstrated the emerging involvement of transfer RNA (tRNA) processing during the progression of tumours. Nevertheless, the roles and regulating mechanisms of tRNA processing genes in neuroblastoma (NB), the prevalent malignant tumour outside the brain in children, are yet unknown. METHODS: Analysis of multi-omics results was conducted to identify crucial regulators of downstream tRNA processing genes. Co-immunoprecipitation and mass spectrometry methods were utilised to measure interaction between proteins. The impact of transcriptional regulators on expression of downstream genes was measured by dual-luciferase reporter, chromatin immunoprecipitation, western blotting and real-time quantitative reverse transcription-polymerase chain reaction (RT-PCR) methods. Studies have been conducted to reveal impact and mechanisms of transcriptional regulators on biological processes of NB. Survival differences were analysed using the log-rank test. RESULTS: c-Myc was identified as a transcription factor driving tRNA processing gene expression and subsequent malate-aspartate shuttle (MAS) in NB cells. Mechanistically, c-Myc directly promoted the expression of glutamyl-prolyl-tRNA synthetase (EPRS) and leucyl-tRNA synthetase (LARS), resulting in translational up-regulation of glutamic-oxaloacetic transaminase 1 (GOT1) as well as malate dehydrogenase 1 (MDH1) via inhibiting general control nonrepressed 2 or activating mechanistic target of rapamycin signalling. Meanwhile, lamin A (LMNA) inhibited c-Myc transactivation via physical interaction, leading to suppression of MAS, aerobic glycolysis, tumourigenesis and aggressiveness. Pre-clinically, lobeline was discovered as a LMNA-binding compound to facilitate its interaction with c-Myc, which inhibited aminoacyl-tRNA synthetase expression, MAS and tumour progression of NB, as well as growth of organoid derived from c-Myc knock-in mice. Low levels of LMNA or elevated expression of c-Myc, EPRS, LARS, GOT1 or MDH1 were linked to a worse outcome and a shorter survival time of clinical NB patients. CONCLUSIONS: These results suggest that targeting c-Myc transactivation by LMNA inhibits tRNA processing essential for MAS and tumour progression.


Subject(s)
Proto-Oncogene Proteins c-myc , Humans , Mice , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Animals , Aspartic Acid/metabolism , Malates/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Neuroblastoma/metabolism , Neuroblastoma/genetics , Disease Progression , Transcriptional Activation/genetics , Cell Line, Tumor , Disease Models, Animal
5.
J Immunother Cancer ; 12(5)2024 May 23.
Article in English | MEDLINE | ID: mdl-38782540

ABSTRACT

BACKGROUND: Approximately half of the neuroblastoma patients develop high-risk neuroblastoma. Current treatment involves a multimodal strategy, including immunotherapy with dinutuximab (IgG ch14.18) targeting GD2. Despite achieving promising results, the recurrence rate remains high and poor survival persists. The therapeutic efficacy of dinutuximab is compromised by suboptimal activation of neutrophils and severe neuropathic pain, partially induced by complement activation. METHODS: To enhance neutrophil cytotoxicity, IgG ch14.18 was converted to the IgA isotype, resulting in potent neutrophil-mediated antibody-dependent cell-mediated cytotoxicity (ADCC), without complement activation. However, myeloid checkpoint molecules hamper neutrophil cytotoxicity, for example through CD47 that is overexpressed on neuroblastomas and orchestrates an immunosuppressive environment upon ligation to signal regulatory protein alpha (SIRPα) expressed on neutrophils. In this study, we combined IgA therapy with CD47 blockade. RESULTS: In vitro killing assays showed enhanced IgA-mediated ADCC by neutrophils targeting neuroblastoma cell lines and organoids in comparison to IgG. Notably, when combined with CD47 blockade, both IgG and IgA therapy were enhanced, though the combination with IgA resulted in the greatest improvement of ADCC. Furthermore, in a neuroblastoma xenograft model, we systemically blocked CD47 with a SIRPα fusion protein containing an ablated IgG1 Fc, and compared IgA therapy to IgG therapy. Only IgA therapy combined with CD47 blockade increased neutrophil influx to the tumor microenvironment. Moreover, the IgA combination strategy hampered tumor outgrowth most effectively and prolonged tumor-specific survival. CONCLUSION: These promising results highlight the potential to enhance immunotherapy efficacy against high-risk neuroblastoma through improved neutrophil cytotoxicity by combining IgA therapy with CD47 blockade.


Subject(s)
CD47 Antigen , Immunoglobulin A , Neuroblastoma , Neutrophils , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/metabolism , CD47 Antigen/immunology , Humans , Neuroblastoma/immunology , Neuroblastoma/drug therapy , Neutrophils/immunology , Neutrophils/metabolism , Animals , Mice , Immunoglobulin A/immunology , Immunoglobulin A/pharmacology , Immunoglobulin A/metabolism , Cell Line, Tumor , Antibody-Dependent Cell Cytotoxicity , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Xenograft Model Antitumor Assays , Immunotherapy/methods , Female , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use
6.
J Cell Mol Med ; 28(10): e18360, 2024 May.
Article in English | MEDLINE | ID: mdl-38785199

ABSTRACT

Neuroblastoma (NB), a common solid tumour in young children originating from the sympathetic nervous system during embryonic development, poses challenges despite therapeutic advances like high-dose chemotherapy and immunotherapy. Some survivors still grapple with severe side effects and drug resistance. The role of lncRNA NUTM2A-AS1 has been explored in various cancers, but its function in drug-resistant NB progression is unclear. Our study found that NUTM2A-AS1 expression in cisplatin-resistant NB cells increased in a time- and dose-dependent manner. Knockdown of NUTM2A-AS1 significantly improved NB cell sensitivity to cisplatin and inhibited metastatic abilities. Additionally, we identified B7-H3, an immune checkpoint-related protein, as a NUTM2A-AS1-associated protein in NB cells. NUTM2A-AS1 was shown to inhibit the protein degradation of B7-H3. Moreover, NUTM2A-AS1 modulated immune evasion in cisplatin-resistant NB cells through B7-H3. Furthermore, NUTM2A-AS1 expression in cisplatin-resistant NB cells was transactivated by NR1D1. In summary, our results unveil the molecular or biological relationship within the NR1D1/NUTM2A-AS1/B7-H3 axis in NB cells under cisplatin treatment, providing an intriguing avenue for fundamental research into cisplatin-resistant NB.


Subject(s)
B7 Antigens , Cisplatin , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Neuroblastoma , RNA, Long Noncoding , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Drug Resistance, Neoplasm/genetics , B7 Antigens/metabolism , B7 Antigens/genetics , RNA, Long Noncoding/genetics , Cisplatin/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Immune Evasion , Animals , Proteolysis/drug effects , Mice
7.
Khirurgiia (Mosk) ; (5): 152-160, 2024.
Article in Russian | MEDLINE | ID: mdl-38785252

ABSTRACT

This review is devoted to surgical approach for neurogenic tumors in children. The authors discuss epidemiological data, history of surgical approaches, preoperative imaging and risk factors. A special attention is paid to the influence of surgical interventions for various neuroblastomas on overall and event-free survival in pediatric population, as well as the most common surgical complications and modern approaches to their treatment.


Subject(s)
Abdominal Neoplasms , Neuroblastoma , Humans , Neuroblastoma/surgery , Child , Abdominal Neoplasms/surgery , Postoperative Complications/etiology , Postoperative Complications/epidemiology , Postoperative Complications/prevention & control , Risk Factors
8.
FASEB J ; 38(10): e23689, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38785406

ABSTRACT

Neuroblastoma, a prevalent extracranial solid tumor in children, arises from undifferentiated nerve cells. While tumor vasculature, often characterized by increased permeability, influences metastasis and recurrence, the direct impact of blood-borne molecules on tumor progression remains unclear. In the present study, we focused on the effect of exposure to albumin, one of the most abundant proteins in the serum, on human neuroblastoma cells. Albumin exposure elevated oxidative stress and led to mitochondria dysfunction via the activation of TGFß and PI3K pathways, accompanied by an increase in the metastatic and invasive properties of neuroblastoma cells. Proteins relevant to the induction of autophagy were upregulated in response to prolonged albumin exposure. Additionally, pre-exposure to albumin before treatment resulted in increased resistance to paclitaxel. Two valeriana-type iridoid glycosides, patrisophoroside and patrinalloside, recently isolated from Nardostachys jatamansi significantly mitigated the effect of albumin on oxidative stress, cell invasiveness, and chemoresistance. These findings illuminate the potential role of blood-borne molecules, such as albumin, in the progression and metastasis of neuroblastoma, as well as the possible therapeutic implications of valeriana-type iridoid glycosides in anti-cancer treatment.


Subject(s)
Drug Resistance, Neoplasm , Iridoid Glycosides , Neuroblastoma , Paclitaxel , Humans , Neuroblastoma/pathology , Neuroblastoma/metabolism , Neuroblastoma/drug therapy , Drug Resistance, Neoplasm/drug effects , Paclitaxel/pharmacology , Iridoid Glycosides/pharmacology , Cell Line, Tumor , Neoplasm Invasiveness , Oxidative Stress/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Valerian/chemistry , Serum Albumin/metabolism
9.
Pediatr Blood Cancer ; 71(7): e31039, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38689540

ABSTRACT

BACKGROUND: Opsoclonus-myoclonus-ataxia syndrome (OMAS) is a rare autoimmune disorder of the nervous system presenting with abnormal eye and limb movements, altered gait, and increased irritability. Two to four percent of children diagnosed with neuroblastoma have neuroblastoma-associated OMAS (NA-OMAS). These children typically present with non-high-risk neuroblastoma that is cured with surgery, with or without chemotherapy. Despite excellent overall survival, patients with NA-OMAS can have significant persistent neurological and developmental issues. OBJECTIVE: This study aimed to describe long-term neurocognitive and adaptive functioning of patients with NA-OMAS treated with multimodal therapy, including intravenous immunoglobulin (IVIG) on Children's Oncology Group (COG) protocol ANBL00P3. METHODS: Of 53 children enrolled on ANBL00P3, 25 submitted evaluable neurocognitive data at diagnosis and at least one additional time point within 2 years and were included in the analyses. Adaptive development was assessed via the Vineland Adaptive Behavior Scale, and validated, age-appropriate measures of intellectual function were also administered. RESULTS: Twenty-one of the 25 patients in this cohort ultimately received IVIG. Descriptive spaghetti plots suggest that this cohort demonstrated stable long-term cognitive functioning and adaptive development over time. This cohort also demonstrated decreased OMAS scores over time consistent with improved OMAS symptoms. CONCLUSIONS: While statistical significance is limited by small sample size and loss to follow-up over 10 years, findings suggest stable long-term cognitive and adaptive functioning over time in this treated cohort.


Subject(s)
Neuroblastoma , Opsoclonus-Myoclonus Syndrome , Humans , Opsoclonus-Myoclonus Syndrome/therapy , Opsoclonus-Myoclonus Syndrome/etiology , Male , Female , Neuroblastoma/complications , Neuroblastoma/therapy , Neuroblastoma/mortality , Child, Preschool , Child , Infant , Immunoglobulins, Intravenous/therapeutic use , Follow-Up Studies , Adolescent , Combined Modality Therapy , Prognosis , Adaptation, Psychological , Cognition , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
10.
Pediatr Blood Cancer ; 71(7): e31051, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38706187

ABSTRACT

It is not clear whether trial access disparities exist in the Children's Oncology Group (COG). Here, we leverage a cohort of children with high-risk neuroblastoma (HR-NBL) enrolled on the COG ANBL00B1 neuroblastoma biology study to examine subsequent enrollment to upfront COG therapeutic trials by race, ethnicity, and proxied poverty status. Among 1917 children with HR-NBL enrolled on ANBL00B1, 696 (36.3%) subsequently enrolled on an upfront therapeutic trial with no difference by race, ethnicity, or proxied poverty status. In neuroblastoma, trial access disparities are not comparable to adult oncology, and efforts to advance equity should prioritize other mechanisms of survival disparities.


Subject(s)
Neuroblastoma , Poverty , Humans , Neuroblastoma/therapy , Neuroblastoma/ethnology , Male , Female , Child , Child, Preschool , Infant , Ethnicity/statistics & numerical data , Clinical Trials as Topic/statistics & numerical data , Healthcare Disparities , Adolescent , Follow-Up Studies
11.
FASEB J ; 38(10): e23644, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38738472

ABSTRACT

Tumors typically lack canonical danger signals required to activate adaptive immunity and also frequently employ substantial immunomodulatory mechanisms that downregulate adaptive responses and contribute to escape from immune surveillance. Given the variety of mechanisms involved in shielding tumors from immune recognition, it is not surprising that single-agent immunomodulatory approaches have been largely unsuccessful in generating durable antitumor responses. Here we report a unique combination of immunomodulatory and cytostatic agents that recondition the tumor microenvironment and eliminate complex and/or poor-prognosis tumor types including the non-immunogenic 4T-1 model of TNBC, the aggressive MOC-2 model of HNSCC, and the high-risk MYCN-amplified model of neuroblastoma. A course of therapy optimized for TNBC cured a majority of tumors in both ectopic and orthotopic settings and eliminated metastatic spread in all animals tested at the highest doses. Immune responses were transferable between therapeutic donor and naïve recipient through adoptive transfer, and a sizeable abscopal effect on distant, untreated lesions could be demonstrated experimentally. Similar results were observed in HNSCC and neuroblastoma models, with characteristic remodeling of the tumor microenvironment documented in all model systems. scRNA-seq analysis implicated upregulation of innate immune responses and antigen presentation in tumor cells and the myeloid cell compartment as critical early events. This analysis also highlighted the potential importance of the autonomic nervous system in the governance of inflammatory processes. The data indicate that the targeting of multiple pathways and mechanisms of action can result in substantial synergistic antitumor effects and suggest follow-up in the neoadjuvant setting may be warranted.


Subject(s)
Tumor Microenvironment , Animals , Mice , Tumor Microenvironment/immunology , Cell Line, Tumor , Neuroblastoma/immunology , Neuroblastoma/therapy , Neuroblastoma/pathology , Female , Humans , Immunomodulation , Mice, Inbred C57BL
12.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38732012

ABSTRACT

Neuroblastoma (NB) is the most commonly diagnosed extracranial solid tumor in children, accounting for 15% of all childhood cancer deaths. Although the 5-year survival rate of patients with a high-risk disease has increased in recent decades, NB remains a challenge in pediatric oncology, and the identification of novel potential therapeutic targets and agents is an urgent clinical need. The RNA-binding protein LIN28B has been identified as an oncogene in NB and is associated with a poor prognosis. Given that LIN28B acts by negatively regulating the biogenesis of the tumor suppressor let-7 miRNAs, we reasoned that selective interference with the LIN28B/let-7 miRNA interaction would increase let-7 miRNA levels, ultimately leading to reduced NB aggressiveness. Here, we selected (-)-epigallocatechin 3-gallate (EGCG) out of 4959 molecules screened as the molecule with the best inhibitory activity on LIN28B/let-7 miRNA interaction and showed that treatment with PLC/PLGA-PEG nanoparticles containing EGCG (EGCG-NPs) led to an increase in mature let-7 miRNAs and a consequent inhibition of NB cell growth. In addition, EGCG-NP pretreatment reduced the tumorigenic potential of NB cells in vivo. These experiments suggest that the LIN28B/let-7 miRNA axis is a good therapeutic target in NB and that EGCG, which can interfere with this interaction, deserves further preclinical evaluation.


Subject(s)
Catechin , MicroRNAs , Neuroblastoma , RNA-Binding Proteins , Catechin/analogs & derivatives , Catechin/pharmacology , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/metabolism , Neuroblastoma/drug therapy , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Animals , Mice , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Cell Proliferation/drug effects , Xenograft Model Antitumor Assays , Mice, Nude
13.
Int Immunopharmacol ; 133: 112145, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38691920

ABSTRACT

Treatment strategies for paediatric neuroblastoma as well as many other cancers are limited by the unfavourable tumour microenvironment (TME). In this study, the TMEs of neuroblastoma were grouped by their genetic signatures into four distinct subtypes: immune enriched, immune desert, non-proliferative and fibrotic. An Immune Score and a Proliferation Score were constructed based on the molecular features of the subtypes to quantify the immune microenvironment or malignancy degree of cancer cells in neuroblastoma, respectively. The Immune Score correlated with a patient's response to immunotherapy; the Proliferation Score was an independent prognostic biomarker for neuroblastoma and proved to be more accurate than the existing clinical predictors. This double scoring system was further validated and the conserved molecular pattern associated with immune landscape and malignancy degree was confirmed. Axitinib and BI-2536 were confirmed as candidate drugs for neuroblastoma by the double scoring system. Both in vivo and in vitro experiments demonstrated that axitinib-induced pyroptosis of neuroblastoma cells activated anti-tumour immunity and inhibited tumour growth; BI-2536 induced cell cycle arrest at the S phase in neuroblastoma cells. The comprehensive double scoring system of neuroblastoma may predict prognosis and screen for therapeutic strategies which could provide personalized treatments.


Subject(s)
Axitinib , Immunotherapy , Neuroblastoma , Tumor Microenvironment , Neuroblastoma/immunology , Neuroblastoma/therapy , Neuroblastoma/pathology , Neuroblastoma/drug therapy , Humans , Tumor Microenvironment/immunology , Prognosis , Animals , Immunotherapy/methods , Cell Line, Tumor , Axitinib/therapeutic use , Child , Male , Female , Child, Preschool , Mice , Infant , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects
15.
Nat Commun ; 15(1): 3745, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702304

ABSTRACT

Early childhood tumours arise from transformed embryonic cells, which often carry large copy number alterations (CNA). However, it remains unclear how CNAs contribute to embryonic tumourigenesis due to a lack of suitable models. Here we employ female human embryonic stem cell (hESC) differentiation and single-cell transcriptome and epigenome analysis to assess the effects of chromosome 17q/1q gains, which are prevalent in the embryonal tumour neuroblastoma (NB). We show that CNAs impair the specification of trunk neural crest (NC) cells and their sympathoadrenal derivatives, the putative cells-of-origin of NB. This effect is exacerbated upon overexpression of MYCN, whose amplification co-occurs with CNAs in NB. Moreover, CNAs potentiate the pro-tumourigenic effects of MYCN and mutant NC cells resemble NB cells in tumours. These changes correlate with a stepwise aberration of developmental transcription factor networks. Together, our results sketch a mechanistic framework for the CNA-driven initiation of embryonal tumours.


Subject(s)
Cell Differentiation , DNA Copy Number Variations , N-Myc Proto-Oncogene Protein , Neural Crest , Neuroblastoma , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Neural Crest/metabolism , Neural Crest/pathology , Female , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Chromosome Aberrations , Human Embryonic Stem Cells/metabolism , Transcriptome , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
16.
PLoS One ; 19(5): e0303136, 2024.
Article in English | MEDLINE | ID: mdl-38743689

ABSTRACT

Superoxide dismutase (SOD) is an antioxidant enzyme that protects the body from free radicals. It has both antioxidant and immunomodulatory properties, inducing macrophage polarization from M1 to M2. Macrophages, key mediators of the innate immune response, are divided into the M1 (pro-inflammatory) and M2 (anti-inflammatory) subtypes. In this study, we aimed to assess the antioxidant and neuroprotective effects of SOD on nerve cells and its immunomodulatory effects on macrophages. We observed that SOD inhibited the accumulation of reactive oxygen species and enhanced the viability of H2O2-treated nerve cells. Furthermore, SOD reduced the degree of necrosis in nerve cells treated with the conditioned medium from macrophages, which induced inflammation. In addition, SOD promoted the M1 to M2 transition of macrophages. Our findings suggest that SOD protects nerve cells and regulates immune responses.


Subject(s)
Macrophages , Neuroprotective Agents , Reactive Oxygen Species , Superoxide Dismutase , Animals , Superoxide Dismutase/metabolism , Mice , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Humans , Neuroprotective Agents/pharmacology , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Neuroblastoma/immunology , Neuroblastoma/pathology , Cell Line, Tumor , Hydrogen Peroxide/pharmacology , Cell Survival/drug effects , Antioxidants/pharmacology
17.
J Exp Clin Cancer Res ; 43(1): 141, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745192

ABSTRACT

BACKGROUND: Neuroblastoma (NB) patients with amplified MYCN often face a grim prognosis and are resistant to existing therapies, yet MYCN protein is considered undruggable. KAP1 (also named TRIM28) plays a crucial role in multiple biological activities. This study aimed to investigate the relationship between KAP1 and MYCN in NB. METHODS: Transcriptome analyses and luciferase reporter assay identified that KAP1 was a downstream target of MYCN. The effects of KAP1 on cancer cell proliferation and colony formation were explored using the loss-of-function assays in vitro and in vivo. RNA stability detection was used to examine the influence of KAP1 on MYCN expression. The mechanisms of KAP1 to maintain MYCN mRNA stabilization were mainly investigated by mass spectrum, immunoprecipitation, RIP-qPCR, and western blotting. In addition, a xenograft mouse model was used to reveal the antitumor effect of STM2457 on NB. RESULTS: Here we identified KAP1 as a critical regulator of MYCN mRNA stability by protecting the RNA N6-methyladenosine (m6A) reader YTHDC1 protein degradation. KAP1 was highly expressed in clinical MYCN-amplified NB and was upregulated by MYCN. Reciprocally, KAP1 knockdown reduced MYCN mRNA stability and inhibited MYCN-amplified NB progression. Mechanistically, KAP1 regulated the stability of MYCN mRNA in an m6A-dependent manner. KAP1 formed a complex with YTHDC1 and RNA m6A writer METTL3 to regulate m6A-modified MYCN mRNA stability. KAP1 depletion decreased YTHDC1 protein stability and promoted MYCN mRNA degradation. Inhibiting MYCN mRNA m6A modification synergized with chemotherapy to restrain tumor progression in MYCN-amplified NB. CONCLUSIONS: Our research demonstrates that KAP1, transcriptionally activated by MYCN, forms a complex with YTHDC1 and METTL3, which in turn maintain the stabilization of MYCN mRNA in an m6A-dependent manner. Targeting m6A modification by STM2457, a small-molecule inhibitor of METTL3, could downregulate MYCN expression and attenuate tumor proliferation. This finding provides a new alternative putative therapeutic strategy for MYCN-amplified NB.


Subject(s)
N-Myc Proto-Oncogene Protein , Neuroblastoma , Tripartite Motif-Containing Protein 28 , Humans , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Mice , Animals , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Tripartite Motif-Containing Protein 28/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA Stability , Cell Line, Tumor , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Mice, Nude , Adenosine/analogs & derivatives , Adenosine/metabolism
19.
NPJ Syst Biol Appl ; 10(1): 52, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760476

ABSTRACT

Neuroblastoma (NB) is one of the leading causes of cancer-associated death in children. MYCN amplification is a prominent genetic marker for NB, and its targeting to halt NB progression is difficult to achieve. Therefore, an in-depth understanding of the molecular interactome of NB is needed to improve treatment outcomes. Analysis of NB multi-omics unravels valuable insight into the interplay between MYCN transcriptional and miRNA post-transcriptional modulation. Moreover, it aids in the identification of various miRNAs that participate in NB development and progression. This study proposes an integrated computational framework with three levels of high-throughput NB data (mRNA-seq, miRNA-seq, and methylation array). Similarity Network Fusion (SNF) and ranked SNF methods were utilized to identify essential genes and miRNAs. The specified genes included both miRNA-target genes and transcription factors (TFs). The interactions between TFs and miRNAs and between miRNAs and their target genes were retrieved where a regulatory network was developed. Finally, an interaction network-based analysis was performed to identify candidate biomarkers. The candidate biomarkers were further analyzed for their potential use in prognosis and diagnosis. The candidate biomarkers included three TFs and seven miRNAs. Four biomarkers have been previously studied and tested in NB, while the remaining identified biomarkers have known roles in other types of cancer. Although the specific molecular role is yet to be addressed, most identified biomarkers possess evidence of involvement in NB tumorigenesis. Analyzing cellular interactome to identify potential biomarkers is a promising approach that can contribute to optimizing efficient therapeutic regimens to target NB vulnerabilities.


Subject(s)
Biomarkers, Tumor , Computational Biology , Gene Regulatory Networks , MicroRNAs , Neuroblastoma , Neuroblastoma/genetics , Humans , MicroRNAs/genetics , Biomarkers, Tumor/genetics , Gene Regulatory Networks/genetics , Computational Biology/methods , Gene Expression Regulation, Neoplastic/genetics , Transcription Factors/genetics , Gene Expression Profiling/methods , RNA, Messenger/genetics , Multiomics
20.
Cancer Lett ; 591: 216882, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38636893

ABSTRACT

Super enhancers (SEs) are genomic regions comprising multiple closely spaced enhancers, typically occupied by a high density of cell-type-specific master transcription factors (TFs) and frequently enriched in key oncogenes in various tumors, including neuroblastoma (NB), one of the most prevalent malignant solid tumors in children originating from the neural crest. Cyclin-dependent kinase 5 regulatory subunit-associated protein 3 (CDK5RAP3) is a newly identified super-enhancer-driven gene regulated by master TFs in NB; however, its function in NB remains unclear. Through an integrated study of publicly available datasets and microarrays, we observed a significantly elevated CDK5RAP3 expression level in NB, associated with poor patient prognosis. Further research demonstrated that CDK5RAP3 promotes the growth of NB cells, both in vitro and in vivo. Mechanistically, defective CDK5RAP3 interfered with the UFMylation system, thereby triggering endoplasmic reticulum (ER) phagy. Additionally, we provide evidence that CDK5RAP3 maintains the stability of MEIS2, a master TF in NB, and in turn, contributes to the high expression of CDK5RAP3. Overall, our findings shed light on the molecular mechanisms by which CDK5RAP3 promotes tumor progression and suggest that its inhibition may represent a novel therapeutic strategy for NB.


Subject(s)
Cell Cycle Proteins , Gene Expression Regulation, Neoplastic , Neuroblastoma , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/metabolism , Animals , Cell Line, Tumor , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Endoplasmic Reticulum/metabolism , Enhancer Elements, Genetic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Mice , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Proliferation , Mice, Nude , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...