1
|
Bento CA, Arnaud-Sampaio VF, Glaser T, Adinolfi E, Coutinho-Silva R, Ulrich H, Lameu C. P2X7 receptor in macrophage polarization and its implications in neuroblastoma tumor behavior. Purinergic Signal 2024:10.1007/s11302-024-10051-w. [PMID: 39425818 DOI: 10.1007/s11302-024-10051-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/15/2024] [Indexed: 10/21/2024] Open
Abstract
Tumor-associated macrophages (TAMs) exhibit antitumor or protumor responses related to inflammatory (or M1) and alternative (or M2) phenotypes, respectively. The P2X7 receptor plays a key role in macrophage polarization, influencing inflammation and immunosuppression. In this study, we investigated the role of the P2X7 receptor in TAMs. Using P2X7 receptor-deficient macrophages, we analyzed gene expression profiles and their implications for neuroblastoma invasion and chemoresistance. Our results showed that P2X7 receptor deficiency altered the expression of classical polarization markers, such as nitric oxide synthase 2 (Nos2) and tumor necrosis factor-α (Tnf), as well as alternative phenotype markers, including mannose receptor C-type 1 (Mrc1) and arginase 1 (Arg1). P2X7 deficiency also influenced the expression of the ectonucleotidases Entpd1 and Nt5e and other purinergic receptors, especially P2ry2, suggesting compensatory mechanisms involved in macrophage polarization. In particular, TAMs deficient in P2X7 showed a phenotype with characteristics intermideiate between resting macrophages (M0) and M1 polarization rather than the M2-type phenotype like and wild-type TAM macrophages. In addition, P2rx7-/- TAMs regulated the expression of P2X7 receptor isoforms in neuroblastoma cells, with downregulation of the P2X7 A and B isoforms leading to a decrease in chemotherapy-induced cell death. However, TAMs expressing P2X7 downregulated only the B isoform, suggesting that TAMs play a role in modulating tumor behavior through P2X7 receptor isoform regulation. Taken together, our data underscore the regulatory function of the P2X7 receptor in orchestrating alternative macrophage polarization and in the interplay between tumor cells and TAMs. These findings help to clarify the complex interplay of purinergic signaling in cancer progression and open up avenues for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Carolina Adriane Bento
- Metastasis Molecular Mechanisms Laboratory and Neurosciences Laboratory, Institute of Chemistry, Biochemistry Department, University of Sao Paulo, Sao Paulo, Brazil
| | - Vanessa Fernandes Arnaud-Sampaio
- Metastasis Molecular Mechanisms Laboratory and Neurosciences Laboratory, Institute of Chemistry, Biochemistry Department, University of Sao Paulo, Sao Paulo, Brazil
| | - Talita Glaser
- Metastasis Molecular Mechanisms Laboratory and Neurosciences Laboratory, Institute of Chemistry, Biochemistry Department, University of Sao Paulo, Sao Paulo, Brazil
| | - Elena Adinolfi
- Section of General Pathology, Department of Experimental and Diagnostic Medicine, University of Ferrara, Ferrara, Italy
| | - Robson Coutinho-Silva
- Laboratory of Immunophysiology Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Henning Ulrich
- Metastasis Molecular Mechanisms Laboratory and Neurosciences Laboratory, Institute of Chemistry, Biochemistry Department, University of Sao Paulo, Sao Paulo, Brazil
| | - Claudiana Lameu
- Metastasis Molecular Mechanisms Laboratory and Neurosciences Laboratory, Institute of Chemistry, Biochemistry Department, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
2
|
Prathipati P, Pathania AS, Chaturvedi NK, Gupta SC, Byrareddy SN, Coulter DW, Challagundla KB. SAP30, an oncogenic driver of progression, poor survival, and drug resistance in neuroblastoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:101543. [PMID: 38817681 PMCID: PMC11137595 DOI: 10.1016/j.omtn.2022.03.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 03/18/2022] [Indexed: 06/01/2024]
Abstract
Neuroblastoma is the most devastating extracranial solid malignancy in children. Despite an intense treatment regimen, the prognosis for high-risk neuroblastoma patients remains poor, with less than 40% survival. So far, MYCN amplification status is considered the most prognostic factor but corresponds to only ∼25% of neuroblastoma patients. Therefore, it is essential to identify a better prognosis and therapy response marker in neuroblastoma patients. We applied robust bioinformatic data mining tools, such as weighted gene co-expression network analysis, cisTarget, and single-cell regulatory network inference and clustering on two neuroblastoma patient datasets. We found Sin3A-associated protein 30 (SAP30), a driver transcription factor positively associated with high-risk, progression, stage 4, and poor survival in neuroblastoma patient cohorts. Tumors of high-risk neuroblastoma patients and relapse-specific patient-derived xenografts showed higher SAP30 levels. The advanced pharmacogenomic analysis and CRISPR-Cas9 screens indicated that SAP30 essentiality is associated with cisplatin resistance and further showed higher levels in cisplatin-resistant patient-derived xenograft tumor cell lines. Silencing of SAP30 induced cell death in vitro and led to a reduced tumor burden and size in vivo. Altogether, these results indicate that SAP30 is a better prognostic and cisplatin-resistance marker and thus a potential drug target in high-risk neuroblastoma.
Collapse
Affiliation(s)
- Philip Prathipati
- Laboratory of Bioinformatics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan
| | - Anup S. Pathania
- Department of Biochemistry and Molecular Biology & Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Nagendra K. Chaturvedi
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Subash C. Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Siddappa N. Byrareddy
- Department of Biochemistry and Molecular Biology & Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Don W. Coulter
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kishore B. Challagundla
- Department of Biochemistry and Molecular Biology & Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- The Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
3
|
Goswami N, Singh A, Bharadwaj S, Sahoo AK, Singh IK. Targeting neuroblastoma by small-molecule inhibitors of human ALYREF protein: mechanistic insights using molecular dynamics simulations. J Biomol Struct Dyn 2024; 42:1352-1367. [PMID: 37158061 DOI: 10.1080/07391102.2023.2204376] [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: 11/16/2022] [Accepted: 03/30/2023] [Indexed: 05/10/2023]
Abstract
Neuroblastoma is a tumour of the sympathetic nervous system mainly prevalent in children. Many strategies have been employed to target several drug-targetable proteins for the clinical management of neuroblastoma. However, the heterogeneous nature of neuroblastoma presents serious challenges in drug development for its treatment. Albeit numerous medications have been developed to target various signalling pathways in neuroblastoma, the redundant nature of the tumour pathways makes its suppression unsuccessful. Recently, the quest for neuroblastoma therapy resulted in the identification of human ALYREF, a nuclear protein that plays an essential role in tumour growth and progression. Therefore, this study used the structure-based drug discovery method to identify the putative inhibitors targeting ALYREF for the Neuroblastoma treatment. Herein, a library of 119 blood-brain barrier crossing small molecules from the ChEMBL database was downloaded and docked against the predicted binding pocket of the human ALYREF protein. Based on docking scores, the top four compounds were considered for intermolecular interactions and molecular dynamics simulation analysis, which revealed CHEMBL3752986 and CHEMBL3753744 with substantial affinity and stability with the ALYREF. These results were further supported by binding free energies and essential dynamics analysis of the respective complexes. Hence, this study advocates the sorted compounds targeting ALYREF for further in vitro and in vivo assessment to develop a drug against neuroblastoma.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nidhi Goswami
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Delhi, India
- Neuropharmacology and Drug Delivery Laboratory, Department of Zoology, Daulat Ram College, University of Delhi, Delhi, India
| | - Archana Singh
- Department of Botany, Hansraj College, University of Delhi, Delhi, India
| | - Shiv Bharadwaj
- Department of Biotechnology, Institute of Biotechnology, College of Life and Applied Sciences, Yeungnam University, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Amaresh Kumar Sahoo
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Allahabad, Uttar Pradesh, India
| | - Indrakant K Singh
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Delhi, India
- Delhi School of Public Health, Institute of Eminence, University of Delhi, Delhi, India
| |
Collapse
|
4
|
Italia M, Wertheim KY, Taschner-Mandl S, Walker D, Dercole F. Mathematical Model of Clonal Evolution Proposes a Personalised Multi-Modal Therapy for High-Risk Neuroblastoma. Cancers (Basel) 2023; 15:cancers15071986. [PMID: 37046647 PMCID: PMC10093626 DOI: 10.3390/cancers15071986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Neuroblastoma is the most common extra-cranial solid tumour in children. Despite multi-modal therapy, over half of the high-risk patients will succumb. One contributing factor is the one-size-fits-all nature of multi-modal therapy. For example, during the first step (induction chemotherapy), the standard regimen (rapid COJEC) administers fixed doses of chemotherapeutic agents in eight two-week cycles. Perhaps because of differences in resistance, this standard regimen results in highly heterogeneous outcomes in different tumours. In this study, we formulated a mathematical model comprising ordinary differential equations. The equations describe the clonal evolution within a neuroblastoma tumour being treated with vincristine and cyclophosphamide, which are used in the rapid COJEC regimen, including genetically conferred and phenotypic drug resistance. The equations also describe the agents’ pharmacokinetics. We devised an optimisation algorithm to find the best chemotherapy schedules for tumours with different pre-treatment clonal compositions. The optimised chemotherapy schedules exploit the cytotoxic difference between the two drugs and intra-tumoural clonal competition to shrink the tumours as much as possible during induction chemotherapy and before surgical removal. They indicate that induction chemotherapy can be improved by finding and using personalised schedules. More broadly, we propose that the overall multi-modal therapy can be enhanced by employing targeted therapies against the mutations and oncogenic pathways enriched and activated by the chemotherapeutic agents. To translate the proposed personalised multi-modal therapy into clinical use, patient-specific model calibration and treatment optimisation are necessary. This entails a decision support system informed by emerging medical technologies such as multi-region sequencing and liquid biopsies. The results and tools presented in this paper could be the foundation of this decision support system.
Collapse
Affiliation(s)
- Matteo Italia
- Department of Electronic, Information, and Bioengineering, Politecnico di Milano, 20133 Milano, Italy
- Correspondence:
| | - Kenneth Y. Wertheim
- Insigneo Institute for in Silico Medicine, University of Sheffield, Sheffield S10 2TN, UK
- Department of Computer Science, University of Sheffield, Sheffield S10 2TN, UK
- Centre of Excellence for Data Science, Artificial Intelligence, and Modelling, University of Hull, Kingston upon Hull HU6 7RX, UK
- School of Computer Science, University of Hull, Kingston upon Hull HU6 7RX, UK
| | | | - Dawn Walker
- Insigneo Institute for in Silico Medicine, University of Sheffield, Sheffield S10 2TN, UK
- Department of Computer Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Fabio Dercole
- Department of Electronic, Information, and Bioengineering, Politecnico di Milano, 20133 Milano, Italy
| |
Collapse
|
5
|
Du X, Ding L, Huang S, Li F, Yan Y, Tang R, Ding X, Zhu Z, Wang W. Cathepsin L promotes chemresistance to neuroblastoma by modulating serglycin. Front Pharmacol 2022; 13:920022. [PMID: 36133820 PMCID: PMC9484481 DOI: 10.3389/fphar.2022.920022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/21/2022] [Indexed: 11/26/2022] Open
Abstract
Cathepsin L (CTSL), a lysosomal acid cysteine protease, is found to play a critical role in chemosencitivity and tumor progression. However, the potential roles and molecular mechanisms of CTSL in chemoresistance in neuroblastoma (NB) are still unclear. In this study, the correlation between clinical characteristics, survival and CTSL expression were assessed in Versteeg dataset. The chemoresistant to cisplatin or doxorubicin was detected using CCK-8 assay. Western blot was employed to detect the expression of CTSL, multi-drug resistance proteins, autophagy-related proteins and apoptosis-related proteins in NB cells while knocking down CTSL. Lysosome staining was analyzed to access the expression levels of lysosomes in NB cells. The expression of apoptosis markers was analyzed with immunofluorescence. Various datasets were analyzed to find the potential protein related to CTSL. In addition, a subcutaneous tumor xenografts model in M-NSG mice was used to assess tumor response to CTSL inhibition in vivo. Based on the validation dataset (Versteeg), we confirmed that CTSL served as a prognostic marker for poor clinical outcome in NB patients. We further found that the expression level of CTSL was higher in SK-N-BE (2) cells than in IMR-32 cells. Knocking down CTSL reversed the chemoresistance in SK-N-BE (2) cells. Furthermore, combination of CTSL inhibition and chemotherapy potently blocked tumor growth in vivo. Mechanistically, CTSL promoted chemoresistance in NB cells by up-regulating multi-drug resistance protein ABCB1 and ABCG2, inhibiting the autophagy level and cell apoptpsis. Furthermore, we observed six datasets and found that Serglycin (SRGN) expression was positively associated with CTSL expresssion. CTSL could mediate chemoresistance by up-regulating SRGN expression in NB cells and SRGN expression was positively correlated with poor prognosis of NB patients. Taken together, our findings indicate that the CTSL promotes chemoresistance to cisplatin and doxorubicin by up-regulating the expression of multi-drug resistance proteins and inhibiting the autophagy level and cell apoptosis in NB cells. Thus, CTSL may be a therapeutic target for overcoming chemoresistant to cisplatin and doxorubicin in NB patients.
Collapse
Affiliation(s)
- Xiaohuan Du
- Department of Pharmacy, Children’s Hospital of Soochow University, Suzhou, China
| | - Leyun Ding
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Shungen Huang
- Department of Oncology, Children’s Hospital of Soochow University, Suzhou, China
| | - Fang Li
- Department of Pharmacy, Children’s Hospital of Soochow University, Suzhou, China
| | - Yinghui Yan
- Department of Pharmacy, Children’s Hospital of Soochow University, Suzhou, China
| | - Ruze Tang
- Department of Oncology, Children’s Hospital of Soochow University, Suzhou, China
| | - Xinyuan Ding
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- *Correspondence: Wenjuan Wang, ; Xinyuan Ding, ; Zengyan Zhu,
| | - Zengyan Zhu
- Department of Pharmacy, Children’s Hospital of Soochow University, Suzhou, China
- *Correspondence: Wenjuan Wang, ; Xinyuan Ding, ; Zengyan Zhu,
| | - Wenjuan Wang
- Department of Pharmacy, Children’s Hospital of Soochow University, Suzhou, China
- *Correspondence: Wenjuan Wang, ; Xinyuan Ding, ; Zengyan Zhu,
| |
Collapse
|
6
|
Combining APR-246 and HDAC-Inhibitors: A Novel Targeted Treatment Option for Neuroblastoma. Cancers (Basel) 2021; 13:cancers13174476. [PMID: 34503286 PMCID: PMC8431508 DOI: 10.3390/cancers13174476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Preclinical analyses identified APR-246 as a potent treatment option for neuroblastoma. However, a specific mode of action, sufficient biomarkers and promising combination partners are still missing. Here, we analyze the susceptibilities of different entities and relate them to gene expression profiles and previously described biomarkers. We propose a gene signature, consisting of 13 genes, as a novel predictive biomarker. Furthermore, we provide evidence that APR-246 directly targets metabolic weaknesses in neuroblastoma cell lines, thus hampering ROS detoxification. This makes APR-246 suitable to be combined with ROS-inducing HDAC inhibitors, a treatment combination that has not been described for neuroblastoma thus far. Abstract APR-246 (Eprenetapopt/PRIMA-1Met) is a very potent anti-cancer drug in clinical trials and was initially developed as a p53 refolding agent. As an alternative mode of action, the elevation of reactive oxygen species (ROS) has been proposed. Through an in silico analysis, we investigated the responses of approximately 800 cancer cell lines (50 entities; Cancer Therapeutics Response Portal, CTRP) to APR-246 treatment. In particular, neuroblastoma, lymphoma and acute lymphocytic leukemia cells were highly responsive. With gene expression data from the Cancer Cell Line Encyclopedia (CCLE; n = 883) and patient samples (n = 1643) from the INFORM registry study, we confirmed that these entities express low levels of SLC7A11, a previously described predictive biomarker for APR-246 responsiveness. Combining the CTRP drug response data with the respective CCLE gene expression profiles, we defined a novel gene signature, predicting the effectiveness of APR-246 treatment with a sensitivity of 90% and a specificity of 94%. We confirmed the predicted APR-246 sensitivity in 8/10 cell lines and in ex vivo cultures of patient samples. Moreover, the combination of ROS detoxification-impeding APR-246 with approved HDAC-inhibitors, known to elevate ROS, substantially increased APR-246 sensitivity in cell cultures and in vivo in two zebrafish neuroblastoma xenograft models. These data provide evidence that APR-246, in combination with HDAC-inhibitors, displays a novel potent targeted treatment option for neuroblastoma patients.
Collapse
|