1
|
Park G, Baniya MK, Cha EJ, Sim SJ, Choi JS, Chun KS. Domperidone Induces Apoptosis through Suppression of STAT3 Signaling in Human Renal Cancer Caki-2 Cells. J Cancer Prev 2025; 30:24-31. [PMID: 40201024 PMCID: PMC11973461 DOI: 10.15430/jcp.24.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/04/2025] [Accepted: 03/15/2025] [Indexed: 04/10/2025] Open
Abstract
Renal cancer continues to offer a great challenge for its successful therapy today, thus underscoring the need for effective chemotherapeutic agents. In the current study, we explored the anticancer effects of domperidone, a dopamine D2 receptor (DRD2) antagonist, in renal cancer Caki-2 cells. Domperidone induced dose and time-dependent cytotoxic effects in Caki-2 cells, triggering intrinsic apoptosis via the stimulation of the caspase cascade and PARP cleavage. The cytotoxic effect of domperidone was found to be partially DRD2-dependent. Domperidone treatment markedly augmented the production of intracellular reactive oxygen species which induced the cell death of Caki-2 cells. In addition, domperidone suppressed Janus kinase 2 and STAT3 phosphorylation, leading to inhibition of survival and proliferation of these cells. Hence, domperidone can be considered a promising candidate for renal cancer treatment.
Collapse
Affiliation(s)
- Geumi Park
- College of Pharmacy, Keimyung University, Daegu, Korea
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDIhub), Daegu, Korea
| | | | - Eun-Jeong Cha
- College of Pharmacy, Keimyung University, Daegu, Korea
| | - So Jin Sim
- College of Pharmacy, Keimyung University, Daegu, Korea
| | - Joon-Seok Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan, Korea
| | | |
Collapse
|
2
|
Lo Greco MC, Marano G, La Rocca M, Acquaviva G, Milazzotto R, Liardo RLE, Basile A, Foti PV, Palmucci S, David E, Parisi S, Pontoriero A, Pergolizzi S, Spatola C. Latest Advancements in the Management of H3K27M-Mutant Diffuse Intrinsic Pontine Glioma: A Narrative Review. Cancers (Basel) 2025; 17:420. [PMID: 39941789 PMCID: PMC11815860 DOI: 10.3390/cancers17030420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Despite recent advancements in radiotherapy for Diffuse Intrinsic Pontine Glioma (DIPG), the prognosis of this disease remains poor, highlighting the need for new treatment strategies to improve outcomes. Adding stereotactic biopsy to the diagnostic process for children with DIPG has been crucial in improving the management of this disease. Indeed, the discovery of the H3K27M mutation as a key driver of DIPG has led to the development of new drugs that are more effective than traditional ones. These include nimotuzumab (an anti-EGFR drug) and vinorelbine (a semisynthetic vinca alkaloid) in combination, Panobinostat (a histone deacetylase inhibitor), ONC201 (a drug that blocks the dopamine receptor D2 and inactivates Akt and ERK kinases), and chimeric antigen receptor (CAR) T cells. In terms of local therapy, identifying the H3K27M mutation can help us explore how genetic changes affect treatment response, recurrence patterns, and survival. Beyond the time to first recurrence, specific patterns of tumor recurrence, like leptomeningeal spread, can influence treatment plans. For example, radiotherapy can be adjusted in terms of doses and volumes, based on tumor aggressiveness. Because the H3K27M mutation is linked to higher malignancy, a slightly higher dose could be used for the second round of local irradiation. Additionally, irradiating the entire craniospinal axis could help control both local and leptomeningeal disease.
Collapse
Affiliation(s)
- Maria Chiara Lo Greco
- Radiation Oncology Unit, Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy; (G.M.); (M.L.R.); (S.P.); (A.P.); (S.P.)
| | - Giorgia Marano
- Radiation Oncology Unit, Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy; (G.M.); (M.L.R.); (S.P.); (A.P.); (S.P.)
| | - Madalina La Rocca
- Radiation Oncology Unit, Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy; (G.M.); (M.L.R.); (S.P.); (A.P.); (S.P.)
| | - Grazia Acquaviva
- Radiation Oncology Unit, University Hospital Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy; (G.A.); (R.M.); (R.L.E.L.); (C.S.)
| | - Roberto Milazzotto
- Radiation Oncology Unit, University Hospital Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy; (G.A.); (R.M.); (R.L.E.L.); (C.S.)
| | - Rocco Luca Emanuele Liardo
- Radiation Oncology Unit, University Hospital Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy; (G.A.); (R.M.); (R.L.E.L.); (C.S.)
| | - Antonio Basile
- Department of Medical Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (A.B.); (P.V.F.); (S.P.); (E.D.)
- Radiology I Unit, University Hospital Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy
| | - Pietro Valerio Foti
- Department of Medical Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (A.B.); (P.V.F.); (S.P.); (E.D.)
- Radiology I Unit, University Hospital Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy
| | - Stefano Palmucci
- Department of Medical Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (A.B.); (P.V.F.); (S.P.); (E.D.)
- Radiology I Unit, University Hospital Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy
| | - Emanuele David
- Department of Medical Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (A.B.); (P.V.F.); (S.P.); (E.D.)
- Radiology I Unit, University Hospital Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy
| | - Silvana Parisi
- Radiation Oncology Unit, Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy; (G.M.); (M.L.R.); (S.P.); (A.P.); (S.P.)
| | - Antonio Pontoriero
- Radiation Oncology Unit, Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy; (G.M.); (M.L.R.); (S.P.); (A.P.); (S.P.)
| | - Stefano Pergolizzi
- Radiation Oncology Unit, Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy; (G.M.); (M.L.R.); (S.P.); (A.P.); (S.P.)
| | - Corrado Spatola
- Radiation Oncology Unit, University Hospital Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy; (G.A.); (R.M.); (R.L.E.L.); (C.S.)
- Department of Medical Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (A.B.); (P.V.F.); (S.P.); (E.D.)
| |
Collapse
|
3
|
Fang Z, Kong W, Zhao Z, Sun W, Xu G, Clark LH, Sullivan SA, Tran AQ, Zhou CS, Sun D, Zhao L, Wang J, Zhou C, Bae-Jump VL. Asparagus officinalis L. extract exhibits anti-proliferative and anti-invasive effects in endometrial cancer cells and a transgenic mouse model of endometrial cancer. Front Pharmacol 2024; 15:1507042. [PMID: 39697544 PMCID: PMC11653357 DOI: 10.3389/fphar.2024.1507042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024] Open
Abstract
Introduction Endometrial cancer is the most common malignancy of the female reproductive system in the United States. Asparagus officinalis is a versatile, nutrient-dense, low-calorie vegetable that contains various bioactive metabolites that have shown a variety of biologic functions beneficial to health. The metabolites from asparagus officinalis extracts or asparagus officinalis extracts exhibit significant anti-tumorigenic activity in some pre-clinical models of cancer. Methods Endometrial cancer cells were used to study the effects of asparagus officinalis on anti-proliferation, anti-invasion and increased sensitivity to cisplatin, and obese and lean Lkb1 fl/fl p53 fl/fl mouse model of endometrial cancer was used to study the role of asparagus officinalis in tumor growth. Results Treatment with increasing concentrations of Asparagus officinalis extracts significantly inhibited cell proliferation, reduced glycolytic activity, induced cellular stress and apoptosis, caused cell cycle G1 arrest, increased the sensitivity of cells to cisplatin, reduced cell adhesion and invasion, and activation of AMPK and inhibition of the AKT/mTOR and MAPK signaling pathways in endometrial cancer cells. Moreover, asparagus officinalis extracts suppressed cell adhesion and invasion through the modulation of the epithelial-to-mesenchymal transition process. Asparagus officinalis extract treatment for 4 weeks resulted in a significant reduction in tumor growth in Lkb1 fl/fl p53 fl/fl mice under both obese and lean conditions, with a decrease in Ki-67 and vascular endothelial growth factor expression and an increase in Bip expression in endometrial tumors. Discussion These findings provide strong preclinical evidence for the potential therapeutic benefit of asparagus officinalis extract as a novel dietary strategy in the treatment of endometrial cancer. Further clinical trials of dietary intervention of asparagus officinalis or combination with cisplatin in patients with endometrial cancer are warranted.
Collapse
Affiliation(s)
- Ziwei Fang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Healthcare Hospital, Beijing, China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Weimin Kong
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Healthcare Hospital, Beijing, China
| | - Ziyi Zhao
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Healthcare Hospital, Beijing, China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Guangxu Xu
- Department of Gynecology, Shanghai University of Medicine and Health Sciences Affiliated Sixth People’s Hospital South Campus, Shanghai, China
| | - Leslie H. Clark
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Stephanie A. Sullivan
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Arthur-Quan Tran
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Chang-Sheng Zhou
- Shandong Juxinyuan Asparagus Industry Development Research Institute, HeZe, Shandong, China
| | - Delin Sun
- Shandong Juxinyuan Asparagus Industry Development Research Institute, HeZe, Shandong, China
| | - Luyu Zhao
- Shandong Juxinyuan Agricultural Technology Co, LTD., HeZe, Shandong, China
| | - Jiandong Wang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Healthcare Hospital, Beijing, China
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Victoria L. Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
4
|
Joo SH, Chun KS. Therapeutic strategies for colorectal cancer: antitumor efficacy of dopamine D2 receptor antagonists. Toxicol Res 2024; 40:533-540. [PMID: 39345737 PMCID: PMC11436607 DOI: 10.1007/s43188-024-00259-8] [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: 04/17/2024] [Revised: 07/16/2024] [Accepted: 07/26/2024] [Indexed: 10/01/2024] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of death, accounting for more than half a million deaths annually. Even worse, an increasing number of cancer cases are diagnosed yearly, and two and a half million new cancer cases are estimated to be diagnosed in 2035. Some antipsychotic drugs, especially those targeting dopamine receptor (DR) D2, demonstrated anticancer activity. Studies have revealed the potential of DRD2 antagonists as anticancer therapeutics, whether alone or as an adjuvant, in treating breast cancer, lung cancer, and others. Emerging evidences indicate DRD2 is involved in the CRC biology, and the association between DRD2 and CRC could be utilized in treating CRC. This study selected DRD2 antagonists with anticancer activity to elucidate the possibility of DRD2 antagonists as new therapeutics for treating CRC.
Collapse
Affiliation(s)
- Sang Hoon Joo
- College of Pharmacy, Daegu Catholic University, Gyeongsan, 38430 Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu, 42601 Republic of Korea
| |
Collapse
|
5
|
Messa L, Testa C, Carelli S, Rey F, Jacchetti E, Cereda C, Raimondi MT, Ceri S, Pinoli P. Non-Negative Matrix Tri-Factorization for Representation Learning in Multi-Omics Datasets with Applications to Drug Repurposing and Selection. Int J Mol Sci 2024; 25:9576. [PMID: 39273521 PMCID: PMC11394968 DOI: 10.3390/ijms25179576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
The vast corpus of heterogeneous biomedical data stored in databases, ontologies, and terminologies presents a unique opportunity for drug design. Integrating and fusing these sources is essential to develop data representations that can be analyzed using artificial intelligence methods to generate novel drug candidates or hypotheses. Here, we propose Non-Negative Matrix Tri-Factorization as an invaluable tool for integrating and fusing data, as well as for representation learning. Additionally, we demonstrate how representations learned by Non-Negative Matrix Tri-Factorization can effectively be utilized by traditional artificial intelligence methods. While this approach is domain-agnostic and applicable to any field with vast amounts of structured and semi-structured data, we apply it specifically to computational pharmacology and drug repurposing. This field is poised to benefit significantly from artificial intelligence, particularly in personalized medicine. We conducted extensive experiments to evaluate the performance of the proposed method, yielding exciting results, particularly compared to traditional methods. Novel drug-target predictions have also been validated in the literature, further confirming their validity. Additionally, we tested our method to predict drug synergism, where constructing a classical matrix dataset is challenging. The method demonstrated great flexibility, suggesting its applicability to a wide range of tasks in drug design and discovery.
Collapse
Affiliation(s)
- Letizia Messa
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, 20133 Milan, Italy
| | - Carolina Testa
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, 20133 Milan, Italy
| | - Stephana Carelli
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, 20154 Milan, Italy
- Pediatric Clinical Research Center "Fondazione Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Federica Rey
- Pediatric Clinical Research Center "Fondazione Romeo ed Enrica Invernizzi", Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy
| | - Emanuela Jacchetti
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, 20133 Milan, Italy
| | - Cristina Cereda
- Center of Functional Genomics and Rare Diseases, Buzzi Children's Hospital, 20154 Milan, Italy
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, 20133 Milan, Italy
| | - Stefano Ceri
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, 20133 Milan, Italy
| | - Pietro Pinoli
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, 20133 Milan, Italy
| |
Collapse
|
6
|
Sim SJ, Jang JH, Choi JS, Chun KS. Domperidone, a Dopamine Receptor D2 Antagonist, Induces Apoptosis by Inhibiting the ERK/STAT3-Mediated Pathway in Human Colon Cancer HCT116 Cells. Biomol Ther (Seoul) 2024; 32:568-576. [PMID: 38914471 PMCID: PMC11392662 DOI: 10.4062/biomolther.2024.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/11/2024] [Accepted: 04/22/2024] [Indexed: 06/26/2024] Open
Abstract
Colorectal cancer (CRC) continues to demonstrate high incidence and mortality rates, emphasizing that implementing strategic measures for prevention and treatment is crucial. Recently, the dopamine receptor D2 (DRD2), a G protein-coupled receptor, has been reported to play multiple roles in growth of tumor cells. This study investigated the anticancer potential of domperidone, a dopamine receptor D2 antagonist, in HCT116 human CRC cells. Domperidone demonstrated concentration- and time-dependent reductions in cell viability, thereby inducing apoptosis. The molecular mechanism revealed that domperidone modulated the mitochondrial pathway, decreasing mitochondrial Bcl-2 levels, elevating cytosolic cytochrome C expression, and triggering caspase- 3, -7, and -9 cleavage. Domperidone decreased in formation of β-arrestin2/MEK complex, which contributing to inhibition of ERK activation. Additionally, treatment with domperidone diminished JAK2 and STAT3 activation. Treatment of U0126, the MEK inhibitor, resulted in reduced phosphorylation of MEK, ERK, and STAT3 without alteration of JAK2 activation, indicating that domperidone targeted both MEK-ERK-STAT3 and JAK2-STAT3 signaling pathways, respectively. Immunoblot analysis revealed that domperidone also downregulated DRD2 expression. Domperidone-induced reactive oxygen species (ROS) generation and N-acetylcysteine treatment mitigated ROS levels and restored cell viability. An in vivo xenograft study verified the significant antitumor effects of domperidone. These results emphasize the multifaceted anticancer effects of domperidone, highlighting its potential as a promising therapeutic agent for human CRC.
Collapse
Affiliation(s)
- So Jin Sim
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Jeong-Hoon Jang
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Joon-Seok Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
7
|
Yang FF, Zhao TT, Milaneh S, Zhang C, Xiang DJ, Wang WL. Small molecule targeted therapies for endometrial cancer: progress, challenges, and opportunities. RSC Med Chem 2024; 15:1828-1848. [PMID: 38911148 PMCID: PMC11187550 DOI: 10.1039/d4md00089g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/10/2024] [Indexed: 06/25/2024] Open
Abstract
Endometrial cancer (EC) is a common malignancy among women worldwide, and its recurrence makes it a common cause of cancer-related death. Surgery and external radiation, chemotherapy, or a combination of strategies are the cornerstone of therapy for EC patients. However, adjuvant treatment strategies face certain drawbacks, such as resistance to chemotherapeutic drugs; therefore, it is imperative to explore innovative therapeutic strategies to improve the prognosis of EC. With the development of pathology and pathophysiology, several biological targets associated with EC have been identified, including PI3K/Akt/mTOR, PARP, GSK-3β, STAT-3, and VEGF. In this review, we summarize the progress of small molecule targeted therapies in terms of both basic research and clinical trials and provide cases of small molecules combined with fluorescence properties in the clinical applications of integrated diagnosis and treatment. We hope that this review will facilitate the further understanding of the regulatory mechanism governing the dysregulation of oncogenic signaling in EC and provide insights into the possible future directions of targeted therapeutic regimens for EC treatment by developing new agents with fluorescence properties for the clinical applications of integrated diagnosis and treatment.
Collapse
Affiliation(s)
- Fei-Fei Yang
- Yixing People's Hospital Yixing Jiangsu 214200 China
| | - Tian-Tian Zhao
- School of Life Sciences and Health Engineering, Jiangnan University Wuxi 214122 China
| | - Slieman Milaneh
- School of Life Sciences and Health Engineering, Jiangnan University Wuxi 214122 China
- Department of Pharmaceutical and Chemical Industries, Higher Institute of Applied Science and Technology Damascus Syria
| | - Chun Zhang
- School of Life Sciences and Health Engineering, Jiangnan University Wuxi 214122 China
| | - Da-Jun Xiang
- Xishan People's Hospital of Wuxi City Wuxi Jiangsu 214105 China
| | - Wen-Long Wang
- Yixing People's Hospital Yixing Jiangsu 214200 China
- School of Life Sciences and Health Engineering, Jiangnan University Wuxi 214122 China
| |
Collapse
|
8
|
Sirek T, Sirek A, Borawski P, Ryguła I, Król-Jatręga K, Opławski M, Boroń D, Chalcarz M, Ossowski P, Dziobek K, Zmarzły N, Boroń K, Mickiewicz P, Grabarek BO. Expression Profiles of Dopamine-Related Genes and miRNAs Regulating Their Expression in Breast Cancer. Int J Mol Sci 2024; 25:6546. [PMID: 38928253 PMCID: PMC11203454 DOI: 10.3390/ijms25126546] [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: 04/22/2024] [Revised: 05/21/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
This study aimed to assess the expression profile of messenger RNA (mRNA) and microRNA (miRNA) related to the dopaminergic system in five types of breast cancer in Polish women. Patients with five breast cancer subtypes were included in the study: luminal A (n = 130), luminal B (n = 196, including HER2-, n = 100; HER2+, n = 96), HER2+ (n = 36), and TNBC (n = 43); they underwent surgery, during which tumor tissue was removed along with a margin of healthy tissue (control material). The molecular analysis included a microarray profile of mRNAs and miRNAs associated with the dopaminergic system, a real-time polymerase chain reaction preceded by reverse transcription for selected genes, and determinations of their concentration using enzyme-linked immunosorbent assay (ELISA). The conducted statistical analysis showed that five mRNAs statistically significantly differentiated breast cancer sections regardless of subtype compared to control samples; these were dopamine receptor 2 (DRD2), dopamine receptor 3 (DRD3), dopamine receptor 25 (DRD5), transforming growth factor beta 2 (TGF-β-2), and caveolin 2 (CAV2). The predicted analysis showed that hsa-miR-141-3p can regulate the expression of DRD2 and TGF-β-2, whereas hsa-miR-4441 is potentially engaged in the expression regulation of DRD3 and DRD5. In addition, the expression pattern of DRD5 mRNA can also be regulated by has-miR-16-5p. The overexpression of DRD2 and DRD3, with concomitant silencing of DRD5 expression, confirms the presence of dopaminergic abnormalities in breast cancer patients. Moreover, these abnormalities may be the result of miR-141-3P, miR-16-5p, and miR-4441 activity, regulating proliferation or metastasis.
Collapse
Affiliation(s)
- Tomasz Sirek
- Department of Plastic Surgery, Faculty of Medicine, Academia of Silesia, 40-555 Katowice, Poland
- Department of Plastic and Reconstructive Surgery, Hospital for Minimally Invasive and Reconstructive Surgery in Bielsko-Biała, 43-316 Bielsko-Biala, Poland; (A.S.); (K.K.-J.)
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dabrowa Górnicza, Poland; (I.R.); (D.B.); (P.O.); (K.D.); (N.Z.); (K.B.); (P.M.); (B.O.G.)
| | - Agata Sirek
- Department of Plastic and Reconstructive Surgery, Hospital for Minimally Invasive and Reconstructive Surgery in Bielsko-Biała, 43-316 Bielsko-Biala, Poland; (A.S.); (K.K.-J.)
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dabrowa Górnicza, Poland; (I.R.); (D.B.); (P.O.); (K.D.); (N.Z.); (K.B.); (P.M.); (B.O.G.)
| | | | - Izabella Ryguła
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dabrowa Górnicza, Poland; (I.R.); (D.B.); (P.O.); (K.D.); (N.Z.); (K.B.); (P.M.); (B.O.G.)
| | - Katarzyna Król-Jatręga
- Department of Plastic and Reconstructive Surgery, Hospital for Minimally Invasive and Reconstructive Surgery in Bielsko-Biała, 43-316 Bielsko-Biala, Poland; (A.S.); (K.K.-J.)
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dabrowa Górnicza, Poland; (I.R.); (D.B.); (P.O.); (K.D.); (N.Z.); (K.B.); (P.M.); (B.O.G.)
| | - Marcin Opławski
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, 31-826 Kraków, Poland;
- Department of Gynecology and Obstetrics, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University in Kraków, 30-705 Kraków, Poland
| | - Dariusz Boroń
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dabrowa Górnicza, Poland; (I.R.); (D.B.); (P.O.); (K.D.); (N.Z.); (K.B.); (P.M.); (B.O.G.)
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, 31-826 Kraków, Poland;
- Institute of Clinical Science, Skłodowska-Curie Medical University, 00-136 Warszawa, Poland
- Department of Gynecology and Obstetrics, TOMMED Specjalisci od Zdrowia, 40-662 Katowice, Poland
| | - Michał Chalcarz
- Chalcarz Clinic-Aesthetic Surgery, Aesthetic Medicine, 60-001 Poznan, Poland;
- Bieńkowski Medical Center-Plastic Surgery, 85-020 Bydgoszcz, Poland
| | - Piotr Ossowski
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dabrowa Górnicza, Poland; (I.R.); (D.B.); (P.O.); (K.D.); (N.Z.); (K.B.); (P.M.); (B.O.G.)
| | - Konrad Dziobek
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dabrowa Górnicza, Poland; (I.R.); (D.B.); (P.O.); (K.D.); (N.Z.); (K.B.); (P.M.); (B.O.G.)
| | - Nikola Zmarzły
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dabrowa Górnicza, Poland; (I.R.); (D.B.); (P.O.); (K.D.); (N.Z.); (K.B.); (P.M.); (B.O.G.)
| | - Kacper Boroń
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dabrowa Górnicza, Poland; (I.R.); (D.B.); (P.O.); (K.D.); (N.Z.); (K.B.); (P.M.); (B.O.G.)
| | - Patrycja Mickiewicz
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dabrowa Górnicza, Poland; (I.R.); (D.B.); (P.O.); (K.D.); (N.Z.); (K.B.); (P.M.); (B.O.G.)
| | - Beniamin Oskar Grabarek
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dabrowa Górnicza, Poland; (I.R.); (D.B.); (P.O.); (K.D.); (N.Z.); (K.B.); (P.M.); (B.O.G.)
- Department of Molecular, Biology Gyncentrum Fertility Clinic, 40-055 Katowice, Poland
| |
Collapse
|
9
|
Jiang N, Li D, Han Y, Luo ZG, Liu LB. Overexpression of zinc finger DHHC-type containing 1 is associated with poor prognosis and cancer cell growth and metastasis in uterine corpus endometrial carcinoma. Aging (Albany NY) 2024; 16:9784-9812. [PMID: 38848146 PMCID: PMC11210219 DOI: 10.18632/aging.205899] [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: 10/16/2023] [Accepted: 03/09/2024] [Indexed: 06/09/2024]
Abstract
The zinc finger DHHC-type containing 1 (ZDHHC1) gene is implicated in the pathogenesis and progression of various malignant tumors, but its precise involvement in uterine corpus endometrial carcinoma (UCEC) remains unknown. Thus, this study investigated ZDHHC1 expression in UCEC using publicly available TCGA and Xena databases and elucidated the functions and mechanisms of the ZDHHC1 gene in UCEC progression using bioinformatics and in vitro experiments. The correlation between ZDHHC1 expression and prognosis, clinical features, immune cells, and RNA modifications of UCEC was evaluated using nomograms, correlation, ROC, and survival analyses. The impacts of ZDHHC1 overexpression on UCEC progression and mechanisms were explored with bioinformatics and in vitro experiments. Our study revealed that ZDHHC1 expression was significantly downregulated in UCEC and correlated with poor prognosis, cancer diagnosis, clinical stage, age, weight, body mass index, histological subtypes, residual tumor, tumor grade, and tumor invasion. Notably, Cox regression analysis and constructed nomograms showed that downregulated ZDHHC1 expression was a prognostic factor associated with poor prognosis in patients with UCEC. Conversely, above-normal ZDHHC1 expression inhibited the cell growth, cell cycle transition, migration, and invasion of UCEC cells, which may be related to the cell cycle, DNA replication, PI3K-AKT, and other pathways that promote tumor progression. Altered ZDHHC1 expression in UCEC was significantly associated with RNA modifications and the changes in cancer immune cell populations, such as CD56 bright NK cells, eosinophils, Th2 cells, and cell markers. In conclusion, considerably reduced ZDHHC1 expression in UCEC is associated with cancer cell growth, metastasis, poor prognosis, immune infiltration, and RNA modifications, revealing the promising potential of ZDHHC1 as a prognostic marker for UCEC.
Collapse
Affiliation(s)
- Ni Jiang
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Li
- Department of Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Ye Han
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi-Guo Luo
- Department of Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Lu-Bin Liu
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Marrone L, Romano S, Malasomma C, Di Giacomo V, Cerullo A, Abate R, Vecchione MA, Fratantonio D, Romano MF. Metabolic vulnerability of cancer stem cells and their niche. Front Pharmacol 2024; 15:1375993. [PMID: 38659591 PMCID: PMC11039812 DOI: 10.3389/fphar.2024.1375993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Cancer stem cells (CSC) are the leading cause of the failure of anti-tumor treatments. These aggressive cancer cells are preserved and sustained by adjacent cells forming a specialized microenvironment, termed niche, among which tumor-associated macrophages (TAMs) are critical players. The cycle of tricarboxylic acids, fatty acid oxidation path, and electron transport chain have been proven to play central roles in the development and maintenance of CSCs and TAMs. By improving their oxidative metabolism, cancer cells are able to extract more energy from nutrients, which allows them to survive in nutritionally defective environments. Because mitochondria are crucial bioenergetic hubs and sites of these metabolic pathways, major hopes are posed for drugs targeting mitochondria. A wide range of medications targeting mitochondria, electron transport chain complexes, or oxidative enzymes are currently investigated in phase 1 and phase 2 clinical trials against hard-to-treat tumors. This review article aims to highlight recent literature on the metabolic adaptations of CSCs and their supporting macrophages. A focus is provided on the resistance and dormancy behaviors that give CSCs a selection advantage and quiescence capacity in particularly hostile microenvironments and the role of TAMs in supporting these attitudes. The article also describes medicaments that have demonstrated a robust ability to disrupt core oxidative metabolism in preclinical cancer studies and are currently being tested in clinical trials.
Collapse
Affiliation(s)
- Laura Marrone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Simona Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Chiara Malasomma
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Valeria Di Giacomo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Andrea Cerullo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Rosetta Abate
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | | | - Deborah Fratantonio
- Department of Medicine and Surgery, LUM University Giuseppe Degennaro, Bari, Italy
| | - Maria Fiammetta Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
11
|
Paraghamian SE, Qiu J, Hawkins GM, Zhao Z, Sun W, Fan Y, Zhang X, Suo H, Hao T, Prabhu VV, Allen JE, Zhou C, Bae-Jump V. A novel dopamine receptor D2 antagonist (ONC206) potentiates the effects of olaparib in endometrial cancer. Cancer Biol Ther 2023; 24:2202104. [PMID: 37069726 PMCID: PMC10115124 DOI: 10.1080/15384047.2023.2202104] [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: 01/18/2023] [Accepted: 04/10/2023] [Indexed: 04/19/2023] Open
Abstract
Poly ADP-ribose polymerase (PARP) inhibitors are effective therapies for cancer patients with homologous recombination (HR) deficient tumors. The imipridone ONC206 is an orally bioavailable dopamine receptor D2 antagonist and mitochondrial protease ClpP agonist that has anti-tumorigenic effects in endometrial cancer via induction of apoptosis, activation of the integrated stress response and modulation of PI3K/AKT signaling. Both PARP inhibitors and imipridones are being evaluated in endometrial cancer clinical trials but have yet to be explored in combination. In this manuscript, we evaluated the effects of the PARP inhibitor olaparib in combination with ONC206 in human endometrioid endometrial cancer cell lines and in a genetically engineered mouse model of endometrial cancer. Our results showed that simultaneous exposure of endometrial cancer cells to olaparib and ONC206 resulted in synergistic anti-proliferative effects and increased cellular stress and apoptosis in both cell lines, compared to either drug alone. The combination treatment also decreased expression of the anti-apoptotic protein Bcl-2 and reduced phosphorylation of AKT and S6, with greater effects compared to either drug alone. In the transgenic model of endometrial cancer, the combination of olaparib and ONC206 resulted in a more significant reduction in tumor weight in obese and lean mice compared to ONC206 alone or olaparib alone, together with a considerably decreased Ki-67 and enhanced H2AX expression in obese and lean mice. These results suggest that this novel dual therapy may be worthy of further exploration in clinical trials.
Collapse
Affiliation(s)
- Sarah E. Paraghamian
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jianqing Qiu
- Department of Obstetrics and Gynecology, the Second Hospital of Shandong University, Jinan, China
| | - Gabrielle M. Hawkins
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ziyi Zhao
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Obstetrics and Gynecology, the Second Hospital of Shandong University, Jinan, China
| | - Yali Fan
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Xin Zhang
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Hongyan Suo
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Tianran Hao
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Victoria Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
12
|
Shakya R, Byun MR, Joo SH, Chun KS, Choi JS. Domperidone Exerts Antitumor Activity in Triple-Negative Breast Cancer Cells by Modulating Reactive Oxygen Species and JAK/STAT3 Signaling. Biomol Ther (Seoul) 2023; 31:692-699. [PMID: 37899746 PMCID: PMC10616512 DOI: 10.4062/biomolther.2023.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023] Open
Abstract
The lack of molecular targets hampers the treatment of triple-negative breast cancer (TNBC). In this study, we determined the cytotoxicity of domperidone, a dopamine D2 receptor (DRD2) antagonist in human TNBC BT-549 and CAL-51 cells. Domperidone inhibited cell growth in a dose- and time-dependent manner. The annexin V/propidium iodide staining showed that domperidone induced apoptosis. The domperidone-induced apoptosis was accompanied by the generation of mitochondrial superoxide and the down-regulation of cyclins and CDKs. The apoptotic effect of domperidone on TNBC cells was prevented by pre-treatment with Mito-TEMPO, a mitochondria-specific antioxidant. The prevention of apoptosis with Mito-TEMPO even at concentrations as low as 100 nM, implies that the generation of mitochondrial ROS mediated the domperidone-induced apoptosis. Immunoblot analysis showed that domperidone-induced apoptosis occurred through the down-regulation of the phosphorylation of JAK2 and STAT3. Moreover, domperidone downregulated the levels of D2-like dopamine receptors including DRD2, regardless of their mRNA levels. Our results support further development of DRD2 antagonists as potential therapeutic strategy treating TNBC.
Collapse
Affiliation(s)
- Rajina Shakya
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Mi Ran Byun
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Sang Hoon Joo
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Joon-Seok Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| |
Collapse
|
13
|
Chen S, Luo X, Yang B, Zhuang J, Guo J, Zhu Y, Mo J. The combined signatures of G protein-coupled receptor family and immune landscape provide a prognostic and therapeutic biomarker in endometrial carcinoma. J Cancer Res Clin Oncol 2023; 149:14701-14719. [PMID: 37584707 PMCID: PMC10602984 DOI: 10.1007/s00432-023-05270-4] [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: 07/16/2023] [Accepted: 08/09/2023] [Indexed: 08/17/2023]
Abstract
G protein-coupled receptors (GPRs) are one of the largest surface receptor superfamilies, and many of them play essential roles in biological processes, including immune responses. In this study, we aim to construct a GPR- and tumor immune environment (TME-i)-associated risk signature to predict the prognosis of patients with endometrial carcinoma (EC). The GPR score was generated by applying univariate Cox regression and the Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression in succession. This involved identifying the differentially expressed genes (DEGs) in the Cancer Genome Atlas-Uterine Corpus Endometrioid Carcinoma (TCGA-UCEC) cohort. Simultaneously, the CIBERSORT algorithm was applied to identify the protective immune cells for TME score construction. Subsequently, we combined the GPR and TME scores to establish a GPR-TME classifier for conducting clinical prognosis assessments. Various functional annotation algorithms were used to conduct biological process analysis distinguished by GPR-TME subgroups. Furthermore, weighted correlation network analysis (WGCNA) was applied to depict the tumor somatic mutations landscapes. Finally, we compared the immune-related molecules between GPR-TME subgroups and resorted to the Tumor Immune Dysfunction and Exclusion (TIDE) for immunotherapy response prediction. The mRNA and protein expression of GPR-related gene P2RY14 were, respectively, validated by RT-PCR in clinical samples and HPA database. To conclude, our GPR-TME classifier may aid in predicting the EC patients' prognosis and immunotherapy responses.
Collapse
Affiliation(s)
- Shengyue Chen
- Dalian Medical University, Dalian, Liaoning, China
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xukai Luo
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Baicai Yang
- Jiaxing University Affiliated Women and Children Hospital, Jiaxing, China
| | - Jingming Zhuang
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinshuai Guo
- Dalian Medical University, Dalian, Liaoning, China
| | - Yingjie Zhu
- Jiaxing University Affiliated Women and Children Hospital, Jiaxing, China
| | - Jiahang Mo
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Español P, Rovira R, Caruana P, Luna-Guibourg R, Soler C, Teixeira N, Rodríguez F, Gallardo A, Edwards M, Porta O, Gámez M, Sánchez O, Llurba E, Corchero JL, Céspedes MV. Dopamine receptors D1 and D2 show prognostic significance and potential therapeutic applications for endometrial cancer patients. Gynecol Oncol 2023; 176:25-35. [PMID: 37437489 DOI: 10.1016/j.ygyno.2023.06.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 07/14/2023]
Abstract
OBJECTIVE Catecholaminergic signaling has been a target for therapy in different type of cancers. In this work, we characterized the ADRβ2, DRD1 and DRD2 expression in healthy tissue and endometrial tumors to evaluate their prognostic significance in endometrial cancer (EC), unraveling their possible application as an antitumor therapy. METHODS 109 EC patients were included. The expression of the ADRβ2, DRD1 and DRD2 proteins was evaluated by immunohistochemistry and univariate and multivariate analysis to assess their association with clinic-pathological and outcome variables. Finally, HEC1A and AN3CA EC cell lines were exposed to different concentrations of selective dopaminergic agents alone or in combination to study their effects on cellular viability. RESULTS ADRβ2 protein expression was not associated with clinico-pathological parameters or prognosis. DRD1 protein expression was reduced in tumors samples but showed a significant inverse association with tumor size and stage. DRD2 protein expression was significantly associated with non-endometrioid EC, high grade tumors, tumor size, worse disease-free survival (HR = 3.47 (95%CI:1.35-8.88)) and overall survival (HR = 2.98 (95%CI:1.40-6.34)). The DRD1 agonist fenoldopam showed a reduction of cellular viability in HEC1A and AN3CA cells. The exposure to domperidone, a DRD2 antagonist, significantly reduced cell viability compared to the control. Finally, DRD1 agonism and DRD2 antagonism combination induced a significant reduction in cell viability of the AN3CA cells compared to monotherapy, close to being an additive response than a synergistic effect (CI of 1.1 at 0.5% Fa). CONCLUSION DRD1 and DRD2 expression levels showed a significant association with clinico-pathological parameters. Both the combined activation of DRD1 and blockage of DRD2 may form an innovative strategy to inhibit tumor growth in EC.
Collapse
Affiliation(s)
- Pia Español
- Gynecologic and Oncology Peritoneal group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona 08041, Spain; Department of Obstetrics and Gynecology, Hospital Universitari Son Espases, Palma 07120, Spain.
| | - Ramon Rovira
- Gynecologic and Oncology Peritoneal group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona 08041, Spain
| | - Pablo Caruana
- Gynecologic and Oncology Peritoneal group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| | - Rocío Luna-Guibourg
- Gynecologic and Oncology Peritoneal group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona 08041, Spain
| | - Cristina Soler
- Gynecologic and Oncology Peritoneal group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona 08041, Spain
| | - Natalia Teixeira
- Gynecologic and Oncology Peritoneal group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona 08041, Spain
| | - Francisco Rodríguez
- Gynecologic and Oncology Peritoneal group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| | - Alberto Gallardo
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona 08041, Spain
| | - Maria Edwards
- Gynecologic and Oncology Peritoneal group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| | - Oriol Porta
- Department of Obstetrics and Gynecology, University Hospital Mútua Terrassa, Terrassa 08221, Spain
| | - Maria Gámez
- Department of Pharmacy, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| | - Olga Sánchez
- Women and Perinatal Health Research group. Department of Obstetrics and Gynecology. Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; Primary care interventions to prevent maternal and child chronic diseases of Perinatal and developmental origin network (RICORS), Instituto Salud Carlos III, Madrid, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elisa Llurba
- Department of Obstetrics and Gynecology, Hospital de la Santa Creu i Sant Pau, Barcelona 08041, Spain; Women and Perinatal Health Research group. Department of Obstetrics and Gynecology. Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; Primary care interventions to prevent maternal and child chronic diseases of Perinatal and developmental origin network (RICORS), Instituto Salud Carlos III, Madrid, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jose Luis Corchero
- Institut de Biotecnologia i de Biomedicina and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - María Virtudes Céspedes
- Gynecologic and Oncology Peritoneal group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; Primary care interventions to prevent maternal and child chronic diseases of Perinatal and developmental origin network (RICORS), Instituto Salud Carlos III, Madrid, Spain.
| |
Collapse
|
15
|
O'donnell J, Zhao Z, Buckingham L, Hao T, Suo H, Zhang X, Fan Y, John C, Deng B, Shen X, Sun W, Secord AA, Zhou C, Bae-Jump VL. Ipatasertib exhibits anti‑tumorigenic effects and enhances sensitivity to paclitaxel in endometrial cancer in vitro and in vivo. Int J Oncol 2023; 63:103. [PMID: 37503790 PMCID: PMC10552741 DOI: 10.3892/ijo.2023.5551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/14/2023] [Indexed: 07/29/2023] Open
Abstract
Endometrial cancer is the most common gynecologic cancer and one of the only cancers for which incidence and mortality is steadily increasing. Although curable with surgery in the early stages, endometrial cancer presents a significant clinical challenge in the metastatic and recurrent setting with few novel treatment strategies emerging in the past fifty years. Ipatasertib (IPAT) is an orally bioavailable pan‑AKT inhibitor, which targets all three AKT isoforms and has demonstrated anti‑tumor activity in pre‑clinical models, with clinical trials emerging for many cancer types. In the present study, the MTT assay was employed to evaluate the therapeutic efficacy of IPAT or IPAT in combination with paclitaxel (PTX) in endometrial cancer cell lines and primary cultures of endometrial cancer. The effect of IPAT and PTX on the growth of endometrial tumors was evaluated in a transgenic mouse model of endometrial cancer. Apoptosis was assessed using cleaved caspase assays and cellular stress was assessed using ROS, JC1 and tetramethylrhodamine ethyl ester assays. The protein expression levels of markers of apoptosis and cellular stress, and DNA damage were evaluated using western blotting and immunohistochemistry. IPAT significantly inhibited cell proliferation, caused cell cycle G1 phase arrest, and induced cellular stress and mitochondrial apoptosis in a dose dependent manner in human endometrial cancer cell lines. Combined treatment with low doses of IPAT and PTX led to synergistic inhibition of cell proliferation and induction of cleaved caspase 3 activity in the human endometrial cancer cell lines and the primary cultures. Furthermore, IPAT effectively reduced tumor growth, accompanied by decreased protein expression levels of Ki67 and phosphorylation of S6 in the Lkb1fl/flp53fl/fl mouse model of endometrioid endometrial cancer. The combination of IPAT and PTX resulted in increased expression of phosphorylated‑H2AX and KIF14, markers of DNA damage and microtubule dysfunction respectively, as compared with IPAT alone, PTX alone or placebo‑treated mice. The results of the present study provide a biological rationale to evaluate IPAT and the combination of IPAT and PTX in future clinical trials for endometrial cancer.
Collapse
Affiliation(s)
- Jillian O'donnell
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ziyi Zhao
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, P.R. China
| | - Lindsey Buckingham
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tianran Hao
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongyan Suo
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, P.R. China
| | - Xin Zhang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, P.R. China
| | - Yali Fan
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, P.R. China
| | - Catherine John
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Boer Deng
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, P.R. China
| | - Xiaochang Shen
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100006, P.R. China
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Angeles Alvarez Secord
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Duke Cancer Institute, Duke University, Durham, NC 27705
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Victoria L. Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
16
|
Xu G, Zhao Z, Wysham WZ, Roque DR, Fang Z, Sun W, Yin Y, Deng B, Shen X, Zhou C, Bae-Jump V. Orlistat exerts anti-obesity and anti-tumorigenic effects in a transgenic mouse model of endometrial cancer. Front Oncol 2023; 13:1219923. [PMID: 37601677 PMCID: PMC10436609 DOI: 10.3389/fonc.2023.1219923] [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: 05/09/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Among all cancers, endometrial cancer is most strongly associated with obesity, with more than 65% of endometrial cancers attributable to obesity and being overweight. Fatty acid synthase (FAS), a key lipogenic enzyme, is expressed in endometrial cancer tumors and is associated with a worse prognosis for this disease. Orlistat, an FAS inhibitor, is an FDA-approved weight loss medication that has demonstrated anti-tumor activity in a variety of preclinical cancer models. Methods In this study, the Lkb1fl/flp53fl/fl mouse model of endometroid endometrial cancer was exposed to three diet interventions, including a high fat diet (obese), a low fat diet (lean) and switch from a high fat to a low fat diet, and then exposed to orlistat or placebo. Results The mice fed a high-fat diet had significantly increased body weight and tumor weight compared to mice fed a low-fat diet. Switching from a high-fat diet to a low fat diet led to a reduction in mouse weight and suppressed tumor growth, as compared to both the high fat diet and low fat diet groups. Orlistat effectively decreased body weight in obese mice and inhibited tumor growth in obese, lean, and the high fat diet switch to low fat diet mouse groups through induction of apoptosis. Orlistat also showed anti-proliferative activity in nine of 11 primary cultures of human endometrial cancer. Discussion Our findings provide strong evidence that dietary intervention and orlistat have anti-tumor activity in vivo and supports further investigation of orlistat in combination with dietary interventions for the prevention and treatment of endometrial cancer.
Collapse
Affiliation(s)
- Guangxu Xu
- Department of Gynecology, Fengxian Hospital, Southern Medical University, Shanghai, China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ziyi Zhao
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Weiya Z. Wysham
- Division of Gynecologic Oncology, Legacy Medical Group, Portland, OR, United States
| | - Dario R. Roque
- Division of Gynecologic Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Ziwei Fang
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yajie Yin
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Boer Deng
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Xiaochang Shen
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Victoria Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
17
|
Nguyen TM, Ngoc DTM, Choi JH, Lee CH. Unveiling the Neural Environment in Cancer: Exploring the Role of Neural Circuit Players and Potential Therapeutic Strategies. Cells 2023; 12:1996. [PMID: 37566075 PMCID: PMC10417274 DOI: 10.3390/cells12151996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023] Open
Abstract
The regulation of the immune environment within the tumor microenvironment has provided new opportunities for cancer treatment. However, an important microenvironment surrounding cancer that is often overlooked despite its significance in cancer progression is the neural environment surrounding the tumor. The release of neurotrophic factors from cancer cells is implicated in cancer growth and metastasis by facilitating the infiltration of nerve cells into the tumor microenvironment. This nerve-tumor interplay can elicit cancer cell proliferation, migration, and invasion in response to neurotransmitters. Moreover, it is possible that cancer cells could establish a network resembling that of neurons, allowing them to communicate with one another through neurotransmitters. The expression levels of players in the neural circuits of cancers could serve as potential biomarkers for cancer aggressiveness. Notably, the upregulation of certain players in the neural circuit has been linked to poor prognosis in specific cancer types such as breast cancer, pancreatic cancer, basal cell carcinoma, and stomach cancer. Targeting these players with inhibitors holds great potential for reducing the morbidity and mortality of these carcinomas. However, the efficacy of anti-neurogenic agents in cancer therapy remains underexplored, and further research is necessary to evaluate their effectiveness as a novel approach for cancer treatment. This review summarizes the current knowledge on the role of players in the neural circuits of cancers and the potential of anti-neurogenic agents for cancer therapy.
Collapse
Affiliation(s)
- Tuan Minh Nguyen
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (T.M.N.); (D.T.M.N.)
| | - Dinh Thi Minh Ngoc
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (T.M.N.); (D.T.M.N.)
| | - Jung-Hye Choi
- College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Chang-Hoon Lee
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea; (T.M.N.); (D.T.M.N.)
| |
Collapse
|
18
|
Amjadi O, Hedayatizadeh-Omran A, Zaboli E, Ghaffari-Hamedani MM, Janbabaei G, Ahangari G. Dopamine receptors gene overexpression in the microenvironment of invasive gastric cancer and its potential implications. Mol Biol Rep 2023; 50:6529-6542. [PMID: 37330941 DOI: 10.1007/s11033-023-08541-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/19/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND Gastric cancer (GC) is the fifth most common cancer worldwide and the most commonly diagnosed cancer in Iran. The nervous system provides proximity to tumor cells by releasing neurotransmitters such as dopamine and presenting them to the corresponding receptor-bearing tumors. While nerve fibers infiltrate the tumor microenvironment, little is known about the expression levels of dopamine (DA), dopamine receptors (DRs), and catechol-O-methyltransferase (COMT) in GC patients. METHODS DRs and COMT expression were analyzed in 45 peripheral blood mononuclear cells (PBMCs) and 20 paired tumor and adjacent tissue of GC patients by quantitative polymerase chain reaction. DA was measured in plasma specimens using enzyme-linked immunosorbent assay. Protein-protein interaction analysis was carried out to identify GC-related hub genes. RESULTS Increased expression of DRD1-DRD3 was found in tumor specimens compared with adjacent non-cancerous specimens (P < 0.05). A positive correlation was found between DRD1 and DRD3 expression (P = 0.009); DRD2 and DRD3 expression (P = 0.04). Plasma levels of dopamine were significantly lower in patients (1298 pg/ml) than in controls (4651 pg/ml). DRD1-DRD4 and COMT were up-regulated in PBMCs of patients compared with controls (P < 0.0001). Bioinformatic analyses showed 30 hub genes associated with Protein kinase A and extracellular signal-regulated kinase signaling pathways. CONCLUSIONS The findings indicated dysregulation of DRs and COMT mRNA expression in GC and suggest that the brain- gastrointestinal axis may mediate gastric cancer development. Network analysis revealed that combination treatments could be considered for optimizing and improving the precision treatment of GC.
Collapse
Affiliation(s)
- Omolbanin Amjadi
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O. Box: 1497716316, Iran
| | - Akbar Hedayatizadeh-Omran
- Gastrointestinal Cancer Research Center, Non-Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ehsan Zaboli
- Gastrointestinal Cancer Research Center, Non-Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Ghasem Janbabaei
- Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasem Ahangari
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O. Box: 1497716316, Iran.
| |
Collapse
|
19
|
Churchill ML, Holdsworth-Carson SJ, Cowley KJ, Luu J, Simpson KJ, Healey M, Rogers PAW, Donoghue JF. Using a Quantitative High-Throughput Screening Platform to Identify Molecular Targets and Compounds as Repurposing Candidates for Endometriosis. Biomolecules 2023; 13:965. [PMID: 37371546 DOI: 10.3390/biom13060965] [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: 03/27/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Endometriosis, defined as the growth of hormonally responsive endometrial-like tissue outside of the uterine cavity, is an estrogen-dependent, chronic, pro-inflammatory disease that affects up to 11.4% of women of reproductive age and gender-diverse people with a uterus. At present, there is no long-term cure, and the identification of new therapies that provide a high level of efficacy and favourable long-term safety profiles with rapid clinical access are a priority. In this study, quantitative high-throughput compound screens of 3517 clinically approved compounds were performed on patient-derived immortalized human endometrial stromal cell lines. Following assay optimization and compound criteria selection, a high-throughput screening protocol was developed to enable the identification of compounds that interfered with estrogen-stimulated cell growth. From these screens, 23 novel compounds were identified, in addition to their molecular targets and in silico cell-signalling pathways, which included the neuroactive ligand-receptor interaction pathway, metabolic pathways, and cancer-associated pathways. This study demonstrates for the first time the feasibility of performing large compound screens for the identification of new translatable therapeutics and the improved characterization of endometriosis molecular pathophysiology. Further investigation of the molecular targets identified herein will help uncover new mechanisms involved in the establishment, symptomology, and progression of endometriosis.
Collapse
Affiliation(s)
- Molly L Churchill
- Gynaecology Research Centre, Department of Obstetrics and Gynaecology, University of Melbourne and The Royal Women's Hospital, Parkville, VIC 3052, Australia
| | - Sarah J Holdsworth-Carson
- Gynaecology Research Centre, Department of Obstetrics and Gynaecology, University of Melbourne and The Royal Women's Hospital, Parkville, VIC 3052, Australia
- Julia Argyrou Endometriosis Centre, Epworth HealthCare, Richmond, VIC 3121, Australia
| | - Karla J Cowley
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Parkville, VIC 3010, Australia
| | - Jennii Luu
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Parkville, VIC 3010, Australia
| | - Kaylene J Simpson
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Parkville, VIC 3010, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Martin Healey
- Gynaecology Research Centre, Department of Obstetrics and Gynaecology, University of Melbourne and The Royal Women's Hospital, Parkville, VIC 3052, Australia
- Gynaecology Endometriosis and Pelvic Pain Unit, Royal Women's Hospital, Parkville, VIC 3052, Australia
| | - Peter A W Rogers
- Gynaecology Research Centre, Department of Obstetrics and Gynaecology, University of Melbourne and The Royal Women's Hospital, Parkville, VIC 3052, Australia
| | - J F Donoghue
- Gynaecology Research Centre, Department of Obstetrics and Gynaecology, University of Melbourne and The Royal Women's Hospital, Parkville, VIC 3052, Australia
| |
Collapse
|
20
|
The Role of a Natural Amphibian Skin-Based Peptide, Ranatensin, in Pancreatic Cancers Expressing Dopamine D2 Receptors. Cancers (Basel) 2022; 14:cancers14225535. [PMID: 36428628 PMCID: PMC9688159 DOI: 10.3390/cancers14225535] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
Despite the progress in early diagnostic and available treatments, pancreatic cancer remains one of the deadliest cancers. Therefore, there is an urgent need for novel anticancer agents with a good safety profile, particularly in terms of possible side-effects. Recently dopaminergic receptors have been widely studied as they were proven to play an important role in cancer progression. Although various synthetic compounds are known for their interactions with the dopaminergic system, peptides have recently made a great comeback. This is because peptides are relatively safe, easy to correct in terms of the improvement of their physicochemical and biological properties, and easy to predict. This paper aims to evaluate the anticancer activity of a naturally existing peptide-ranatensin, toward three different pancreatic cancer cell lines. Additionally, since there is no sufficient information confirming the exact character of the interaction between ranatensin and dopaminergic receptors, we provide, for the first time, binding properties of the compound to such receptors.
Collapse
|
21
|
Geng J, Zhang Y, Meng Q, Yan H, Wang Y. The role of liver kinase B1 in tumor progression through regulation of lipid metabolism. Clin Transl Oncol 2022; 24:2045-2054. [PMID: 35896782 PMCID: PMC9522762 DOI: 10.1007/s12094-022-02863-2] [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: 03/12/2022] [Accepted: 05/19/2022] [Indexed: 10/30/2022]
Abstract
The somatic mutation of liver kinase B1 (LKB1) has been implicated in various tumors, which is reflected in the survival, proliferation, and metastasis of tumor cells. However, the regulation of LKB1 in lipid metabolism, a process that is involved in tumor progression is not completely clear. We conclude that LKB1 deficiency results in abnormal expression and activation of multiple molecules related to lipid metabolism which locate downstream of AMP-activated protein kinase (AMPK) or salt-induced kinase (SIK). Abnormal lipid metabolism induced by LKB1 deficiency contributes to the proliferation and metastasis of tumor cells through energy regulation.
Collapse
Affiliation(s)
- Jialu Geng
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Qingfei Meng
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Hang Yan
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China.
| |
Collapse
|
22
|
Zhang W, Li L, Li J, Yu H, Zheng F, Yan B, Cai W, Chen Y, Yin L, Tang D, Xu Y, Dai Y. Systematic Analysis of Neurotransmitter Receptors in Human Breast Cancer Reveals a Strong Association With Outcome and Uncovers HTR6 as a Survival-Associated Gene Potentially Regulating the Immune Microenvironment. Front Immunol 2022; 13:756928. [PMID: 35359970 PMCID: PMC8960964 DOI: 10.3389/fimmu.2022.756928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 02/16/2022] [Indexed: 11/16/2022] Open
Abstract
Many epidemiological reports have indicated an increase in the incidence of breast cancer among psychotic patients, suggesting that the targets of antipsychotics, neurotransmitter receptors, may have a role in tumorigenesis. However, the functions of neurotransmitter receptors in cancer are barely known. Here, we analyzed 44 neurotransmitter receptors in breast cancer and revealed that the expression of 34 receptors was positively correlated with relapse-free survival rates (RFS) of patients using the public database (n = 3951). Among all these receptors, we revealed decreased expression of HTR6 in human advanced breast cancer versus tumors in situ using our original data (n = 44). After a pan-cancer analysis including 22 cancers (n = 11262), we disclosed that HTR6 was expressed in 12 tumors and uncovered its influence on survival in seven tumors. Using multi-omics datasets from Linkedomics, we revealed a potential regulatory role of HTR6 in MAPK, JUN, and leukocyte-differentiation pathways through enriching 294 co-expressed phosphorylated proteins of HTR6. Furthermore, we proclaimed a close association of HTR6 expression with the immune microenvironment. Finally, we uncovered two possible reasons for HTR6 down-regulation in breast cancer, including deep deletion in the genome and the up-regulation of FOXA1 in breast cancer, which was a potential negatively regulatory transcription factor of HTR6. Taken together, we revealed a new function of neurotransmitter receptors in breast cancer and identified HTR6 as a survival-related gene potentially regulating the immune microenvironment. The findings in our study would improve our understanding of the pathogenesis of breast cancer and provided a theoretical basis for personalized medication in psychotic patients.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China.,South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China.,HaploX Biotechnology, Shenzhen, China
| | - Lintai Li
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China.,South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Jianxuan Li
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China.,South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Haiyan Yu
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China.,South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Fengping Zheng
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Bin Yan
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Wanxia Cai
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Yumei Chen
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Lianghong Yin
- Department of Nephrology, Institute of Nephrology and Blood Purifification, The First Affifiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Donge Tang
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Yong Xu
- Shenzhen Second People's Hospital, The First Affifiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yong Dai
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| |
Collapse
|
23
|
Fan Y, Wang J, Fang Z, Pierce SR, West L, Staley A, Tucker K, Yin Y, Sun W, Kong W, Prabhu V, Allen JE, Zhou C, Bae-Jump VL. Anti-Tumor and Anti-Invasive Effects of ONC201 on Ovarian Cancer Cells and a Transgenic Mouse Model of Serous Ovarian Cancer. Front Oncol 2022; 12:789450. [PMID: 35372029 PMCID: PMC8970020 DOI: 10.3389/fonc.2022.789450] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/22/2022] [Indexed: 12/26/2022] Open
Abstract
ONC201 is a promising first-in-class small molecule that has been reported to have anti-neoplastic activity in various types of cancer through activation of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) as well as activation of mitochondrial caseinolytic protease P (ClpP). The present study was to explore the anti-tumor potential effect of ONC201 in ovarian cancer cell lines and in a transgenic mouse model of high grade serous ovarian cancer under obese (high fat diet) and lean (low fat diet) conditions. ONC201 significantly suppressed cell proliferation, induced arrest in G1 phase, and increased cellular stress and apoptosis, accompanied by dual inhibition of the AKT/mTOR/S6 and MAPK pathways in OC cells. ONC201 also resulted in inhibition of adhesion and invasion via epithelial–mesenchymal transition and reduction of VEGF expression. Pre-treatment with the anti-oxidant, N-acetylcysteine (NAC), reversed the ONC201-induced oxidative stress response, and prevented ONC201-reduced VEGF and cell invasion by regulating epithelial–mesenchymal transition protein expression. Knockdown of ClpP in ovarian cancer cells reduced ONC201 mediated the anti-tumor activity and cellular stress. Diet-induced obesity accelerated ovarian tumor growth in the KpB mouse model. ONC201 significantly suppressed tumor growth, and decreased serum VEGF production in obese and lean mice, leading to a decrease in tumoral expression of Ki-67, VEGF and phosphorylation of p42/44 and S6 and an increase in ClpP and DRD5, as assessed by immunohistochemistry. These results suggest that ONC201 may be a promising therapeutic agent to be explored in future clinical trials in high-grade serous ovarian cancer.
Collapse
Affiliation(s)
- Yali Fan
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jiandong Wang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Ziwei Fang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Stuart R. Pierce
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lindsay West
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Allison Staley
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Katherine Tucker
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yajie Yin
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Weimin Kong
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | | | | | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- *Correspondence: Victoria L. Bae-Jump, ; Chunxiao Zhou,
| | - Victoria L. Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- *Correspondence: Victoria L. Bae-Jump, ; Chunxiao Zhou,
| |
Collapse
|
24
|
Tucker K, Yin Y, Staley SA, Zhao Z, Fang Z, Fan Y, Zhang X, Suo H, Sun W, Prabhu VV, Allen JE, Zhou C, Bae-Jump VL. ONC206 has anti-tumorigenic effects in human ovarian cancer cells and in a transgenic mouse model of high-grade serous ovarian cancer. Am J Cancer Res 2022; 12:521-536. [PMID: 35261784 PMCID: PMC8900003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/08/2022] [Indexed: 06/14/2023] Open
Abstract
ONC206, a dopamine receptor D2 (DRD2) antagonist and imipridone, is a chemically modified derivative of ONC201. Recently, ONC206 and other imipridones were identified as activators of the mitochondrial protease ClpP, inducing downstream pathways that allow them to selectively target cancer cells. Clinical trials showed that ONC201, the first in class imipridone, was well tolerated and exhibited tumor regression in some solid tumors. Our goal was to evaluate the effect of ONC206 on cell proliferation and tumor growth in ovarian cancer cell lines and in a transgenic mouse model of high grade serous ovarian cancer (KpB model). ONC206 was more potent than ONC201 in inhibiting cell proliferation, as evidenced by a 10-fold decrease in IC50 for the SKOV3 and OVCAR5 cell lines. This was accompanied by the results that ONC206 significantly inhibited cellular proliferation, induced cell cycle G1 arrest and apoptosis, caused cellular stress, and inhibited adhesion and invasion in vitro. Treatment of obese and non-obese KpB mice with ONC206 elevated Bip and ClpP expression and reduced KI67, BCL-XL and DRD2 expression in the ovarian tumors. Our findings demonstrate that ONC206 has anti-tumorigenic effects in ovarian cancer as previously demonstrated by ONC201 but appears to be as well tolerated and more potent. Thus, ONC206 deserves further evaluation in clinical trials.
Collapse
Affiliation(s)
- Katherine Tucker
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Yajie Yin
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Stuart-Allison Staley
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Rocky Mountain Gynecologic Oncology, Swedish Medical CenterDenver, CO 80113, USA
| | - Ziyi Zhao
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing 100025, China
| | - Ziwei Fang
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing 100025, China
| | - Yali Fan
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing 100025, China
| | - Xin Zhang
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing 100025, China
| | - Hongyan Suo
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing 100025, China
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | | | | | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Victoria L Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| |
Collapse
|
25
|
Staley A, Tucker K, Yin Y, Zhang X, Fan Y, Zhang Y, Fang Z, Sun W, Suo H, Zhao X, Zhao Z, Prabhu VV, Allen JE, Zhou C, Bae-Jump VL. Highly potent dopamine receptor D2 antagonist ONC206 demonstrates anti-tumorigenic activity in endometrial cancer. Am J Cancer Res 2021; 11:5374-5387. [PMID: 34873466 PMCID: PMC8640798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/17/2021] [Indexed: 06/13/2023] Open
Abstract
Endometrial cancer (EC) is a highly obesity-driven cancer, with limited treatment options. ONC201 is an imipridone that selectively antagonizes the G protein-coupled receptors dopamine receptor D2 and D3 (DRD2/3) and activates human mitochondrial caseinolytic protease P (ClpP). It is a promising first-in-class small molecule that has been reported to have anti-neoplastic activity in various types of cancer through induction of the integrated stress response (ISR) as well as through stimulation of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and subsequent induction of apoptosis. ONC201 is being evaluated in Phase II clinical trials for solid tumors and hematological malignancies, including EC. ONC206 is an analog of ONC201 with nanomolar potency in Phase I clinical trials. This study evaluated the anti-tumor efficacy of ONC206 in EC cell lines and the Lkb1fl/flp53fl/fl genetically engineered mouse model of endometrioid EC. ONC206 revealed greater potency than ONC201 in the inhibition of proliferation in EC cell lines, with IC50 concentration ranges of 0.21-0.32 µM for ONC026 versus 2.14-3.53 µM for ONC201. ONC206 induced cellular stress, apoptosis and cell cycle G1 arrest, accompanied by inhibition of the AKT/mTOR/S6 pathways in EC cells. Diet-induced obesity accelerated tumor growth in Lkb1fl/flp53fl/fl mice. ONC206 inhibited EC tumor size and weight in both obese and lean mice after 4 weeks of treatment. Treatment with ONC206 led to a decrease in expression of Ki67, BCL-XL and phosphorylation of S6, as well as an increase in ClpP in endometrial tumors under both obese and lean conditions. Overall, the pre-clinical efficacy of ONC206 is promising and worthy of further exploration in clinical trials for endometrioid EC.
Collapse
Affiliation(s)
- Allison Staley
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Katherine Tucker
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Yajie Yin
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Xin Zhang
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing 100069, China
| | - Yali Fan
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing 100069, China
| | - Yingao Zhang
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Ziwei Fang
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing 100069, China
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Hongyan Suo
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing 100069, China
| | - Xiaoling Zhao
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing 100069, China
| | - Ziyi Zhao
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing 100069, China
| | | | | | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| | - Victoria L Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC 27599, USA
| |
Collapse
|
26
|
Exploring the Molecular Mechanism of Astragali Radix-Curcumae Rhizoma against Gastric Intraepithelial Neoplasia by Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8578615. [PMID: 34646329 PMCID: PMC8505068 DOI: 10.1155/2021/8578615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/22/2021] [Indexed: 01/30/2023]
Abstract
Background Astragali Radix-Curcumae Rhizoma (ARCR), a classic drug pair, has been widely used for the treatment of gastric intraepithelial neoplasia (GIN) in China. However, the underlying mechanisms of this drug pair are still unknown. Thus, elucidating the molecular mechanism of ARCR for treating GIN is imperative. Methods The active components and targets of ARCR were determined from the TCMSP database, and the differentially expressed genes related to GIN were identified from the GSE130823 dataset. The protein-protein interaction (PPI) network and ARCR-active component-target-pathway network were constructed by STRING 11.0 and Cytoscape 3.7.2, respectively. In addition, a receiver operating characteristic curve (ROC) was conducted to verify the key targets, and enrichment analyses were performed using R software. Molecular docking was carried out to test the binding capacity between core active components and key targets. Results 31 active components were obtained from ARCR, among which 22 were hit by the 51 targets associated with GIN. Gene Ontology (GO) functional enrichment analysis showed that biological process (BP), molecular function (MF), and cellular component (CC) were most significantly enriched in response to a drug, catecholamine binding, and apical part of the cell, respectively. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated ARCR against GIN through regulation of neuroactive ligand-receptor interaction, nitrogen metabolism, calcium signaling pathway, chemical carcinogenesis-receptor activation, drug metabolism, gap junction, and cancers. In the PPI network, 15 potential targets were identified, of which nine key targets were proven to have higher diagnostic values in ROC. Molecular docking revealed a good binding affinity of active components (quercetin, bisdemethoxycurcumin, and kaempferol) with the corresponding targets (CYP3A4, CYP1A1, HMOX1, DRD2, DPP4, ADRA2A, ADRA2C, NR1I2, and LGALS4). Conclusion This study revealed the active components and molecular mechanism by which ARCR treatment is effective against GIN through regulating multipathway, such as neuroactive ligand-receptor interaction, nitrogen metabolism, and calcium signaling pathway.
Collapse
|
27
|
Pierce SR, Fang Z, Yin Y, West L, Asher M, Hao T, Zhang X, Tucker K, Staley A, Fan Y, Sun W, Moore DT, Xu C, Tsai YH, Parker J, Prabhu VV, Allen JE, Lee D, Zhou C, Bae-Jump V. Targeting dopamine receptor D2 as a novel therapeutic strategy in endometrial cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:61. [PMID: 33557912 PMCID: PMC7869513 DOI: 10.1186/s13046-021-01842-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/13/2021] [Indexed: 01/04/2023]
Abstract
Background ONC201 is a dopamine receptor D2 (DRD2) antagonist that inhibits tumor growth in preclinical models through ClpP activation to induce integrated stress response pathway and mitochondrial events related to inhibition of cell growth, which is being explored in clinical trials for solid tumors and hematological malignancies. In this study, we investigated the anti-tumorigenic effect of ONC201 in endometrial cancer cell lines and a genetically engineered mouse model of endometrial cancer. Methods Cell proliferation was assessed by MTT and colony formation assays. Cell cycle and apoptosis were evaluated by Cellometer. Invasion capacity was tested using adhesion, transwell and wound healing assays. LKB1fl/flp53fl/fl mouse model of endometrial cancer were fed a control low fat diet versus a high fat diet to mimic diet-induced obesity. Following tumor onset, mice were treated with placebo or ONC201. Metabolomics and lipidomics were used to identify the obesity-dependent effects of ONC201 in the mouse endometrial tumors. DRD2 expression was analyzed by immunohistochemistry in human endometrioid and serous carcinoma specimens. DRD2 mRNA expression from the Cancer Genome Atlas (TCGA) database was compared between the four molecular subtypes of endometrial cancer. Results Increasing DRD2 expression in endometrial cancer was significantly associated with grade, serous histology and stage, as well as worse progression free survival and overall survival. Higher expression of DRD2 mRNA was found for the Copy Number High (CNH) subtype when compared to the other subtypes. ONC201 inhibited cell proliferation, induced cell cycle G1 arrest, caused cellular stress and apoptosis and reduced invasion in endometrial cancer cells. Diet-induced obesity promoted endometrial tumor growth while ONC201 exhibited anti-tumorigenic efficacy in the obese and lean LKB1fl/fl/p53fl/fl mice. Metabolomic analysis demonstrated that ONC201 reversed the obesity-driven upregulation of lipid biosynthesis and reduced protein biosynthesis in obese and lean mice. Conclusion ONC201 has anti-tumorigenic effects in endometrial cancer cells and a transgenic mouse model of endometrial cancer, and DRD2 expression was documented in both human serous and endometrioid endometrial cancer. These studies support DRD2 antagonism via ONC201 as a promising therapeutic strategy for endometrial cancer that has already demonstrated pharmacodynamic activity and clinical benefit in both serous and endometrioid endometrial cancer patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01842-9.
Collapse
Affiliation(s)
- Stuart R Pierce
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ziwei Fang
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yajie Yin
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Lindsay West
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Majdouline Asher
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Tianran Hao
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xin Zhang
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Katherine Tucker
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Allison Staley
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yali Fan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wenchuan Sun
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Dominic T Moore
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chang Xu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yi-Hsuan Tsai
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Joel Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | | | | | | | - Chunxiao Zhou
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Victoria Bae-Jump
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|