1
|
Luan J, Yu M, Gu Q, Zhou X, Shao Y, Chen T, Zhang J, Zhu Z, Song N, Yang J. Fatty acid synthase inhibition improves hypertension-induced erectile dysfunction by suppressing oxidative stress and NLRP3 inflammasome-dependent pyroptosis through activating the Nrf2/HO-1 pathway. Front Immunol 2025; 15:1532021. [PMID: 39877365 PMCID: PMC11772187 DOI: 10.3389/fimmu.2024.1532021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Background Erectile dysfunction (ED) is a prevalent male sexual disorder, commonly associated with hypertension, though the underlying mechanisms remain poorly understood. Objective This study aims to explore the role of Fatty acid synthase (Fasn) in hypertension-induced ED and evaluate the therapeutic potential of the Fasn inhibitor C75. Materials and methods Erectile function was assessed by determining the intracavernous pressure/mean arterial pressure (ICP/MAP) ratio, followed by the collection of cavernous tissue for transcriptomic and non-targeted metabolomic analyses. In vitro, a concentration of 10-6 M angiotensin II (Ang II) was applied to rat aortic endothelial cells (RAOECs) to establish a model of hypertension. In vivo, spontaneously hypertensive rats (SHR) were randomly divided into two groups. The SHR+C75 group received intraperitoneal injections of C75 at a dose of 2 mg/kg once a week. After five weeks of treatment, the erectile function of the rats was assessed, and penile tissues were harvested for further analysis. Molecular and protein expression were assessed using Western blotting, qRT-PCR, immunofluorescence staining, and immunohistochemistry. Results The SHR exhibited ED, indicated by reduced maximum ICP/MAP ratios. Histologically, corpus cavernosum tissue of SHR showed elevated fibrosis and endothelial dysfunction. Additionally, increased expression of the NLRP3 inflammasome, Caspase-1, GSDMD, and the pro-inflammatory cytokines IL-1β and IL-18 was observed. Multi-omics analysis revealed significant enrichment in lipid metabolic pathways, with Fasn identified as a hub gene. In vitro, siFasn and C75 enhanced antioxidant markers Nrf2 and HO-1, reduced ROS accumulation, and suppressed NLRP3 and GSDMD levels. In vivo, C75 treatment restored endothelial function and reversed erectile dysfunction, accompanied by decreased oxidative stress and pyroptosis in the penile corpus cavernosum. Conclusion These findings suggest that Fasn inhibition may offer a promising therapeutic strategy for hypertension-induced ED by alleviating oxidative stress and suppressing NLRP3 inflammasome-dependent endothelial cell pyroptosis via activation of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Jiaochen Luan
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mengchi Yu
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Gu
- Department of Urology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan Zhou
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yunqiang Shao
- Department of Urology, People’s Hospital of Xinjiang Kizilsu Kirgiz Autonomous Prefecture, Kizilsu Kirgiz Autonomous Prefecture, China
| | - Tong Chen
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiayi Zhang
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zheng Zhu
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ninghong Song
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Yang
- Department of Urology, Jiangsu Provincial People’s Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Mo Y, Han Y, Chen Y, Fu C, Li Q, Liu Z, Xiao M, Xu B. ZDHHC20 mediated S-palmitoylation of fatty acid synthase (FASN) promotes hepatocarcinogenesis. Mol Cancer 2024; 23:274. [PMID: 39696259 DOI: 10.1186/s12943-024-02195-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Protein palmitoylation is a reversible fatty acyl modification that undertakes important functions in multiple physiological processes. Dysregulated palmitoylations are frequently associated with the formation of cancer. How palmitoyltransferases for S-palmitoylation are involved in the occurrence and development of hepatocellular carcinoma (HCC) is largely unknown. METHODS Chemical carcinogen diethylnitrosamine (DEN)-induced and DEN combined CCl4 HCC models were used in the zinc finger DHHC-type palmitoyltransferase 20 (ZDHHC20) knockout mice to investigate the role of ZDHHC20 in HCC tumourigenesis. Palmitoylation liquid chromatography-mass spectrometry analysis, acyl-biotin exchange assay, co-immunoprecipitation, ubiquitination assays, protein half-life assays and immunofluorescence microscopy were conducted to explore the downstream regulators and corresponding mechanisms of ZDHHC20 in HCC. RESULTS Knocking out of ZDHHC20 significantly reduced hepatocarcinogenesis induced by chemical agents in the two HCC mouse models in vivo. 97 proteins with 123 cysteine sites were found to be palmitoylated in a ZDHHC20-dependent manner. Among these, fatty acid synthase (FASN) was palmitoylated at cysteines 1471 and 1881 by ZDHHC20. The genetic knockout or pharmacological inhibition of ZDHHC20, as well as the mutation of the critical cysteine sites of FASN (C1471S/C1881S) accelerated the degradation of FASN. Furthermore, ZDHHC20-mediated FASN palmitoylation competed against the ubiquitin-proteasome pathway via the E3 ubiquitin ligase complex SNX8-TRIM28. CONCLUSIONS Our findings demonstrate the critical role of ZDHHC20 in promoting hepatocarcinogenesis, and a mechanism underlying a mutual restricting mode for protein palmitoylation and ubiquitination modifications.
Collapse
Affiliation(s)
- Yaqi Mo
- Center for Intelligent Oncology, Chongqing University Cancer Hospital and Chongqing University School of Medicine, and Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing, 400030, China
| | - Yamei Han
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Yang Chen
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- Department of Radiation Oncology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Chunling Fu
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Qing Li
- Center for Intelligent Oncology, Chongqing University Cancer Hospital and Chongqing University School of Medicine, and Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing, 400030, China
| | - Zhuang Liu
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Mingming Xiao
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Bo Xu
- Center for Intelligent Oncology, Chongqing University Cancer Hospital and Chongqing University School of Medicine, and Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing, 400030, China.
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| |
Collapse
|
3
|
Gao T, Yang L, Zhang Y, Bajinka O, Yuan X. Cancer metabolic reprogramming and precision medicine-current perspective. Front Pharmacol 2024; 15:1450441. [PMID: 39484162 PMCID: PMC11524845 DOI: 10.3389/fphar.2024.1450441] [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: 06/17/2024] [Accepted: 10/04/2024] [Indexed: 11/03/2024] Open
Abstract
Despite the advanced technologies and global attention on cancer treatment strategies, cancer continues to claim lives and adversely affects socio-economic development. Although combination therapies were anticipated to eradicate this disease, the resilient and restorative nature of cancers allows them to proliferate at the expense of host immune cells energetically. This proliferation is driven by metabolic profiles specific to the cancer type and the patient. An emerging field is exploring the metabolic reprogramming (MR) of cancers to predict effective treatments. This mini-review discusses the recent advancements in cancer MR that have contributed to predictive, preventive, and precision medicine. Current perspectives on the mechanisms of various cancer types and prospects for MR and personalized cancer medicine are essential for optimizing metabolic outputs necessary for personalized treatments.
Collapse
Affiliation(s)
- Tingting Gao
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Liuxin Yang
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yali Zhang
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Ousman Bajinka
- School of Medicine and Allied Health Sciences, University of The Gambia, Banjul, Gambia
| | - Xingxing Yuan
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
4
|
Papulino C, Chianese U, Ali A, Favale G, Tuccillo C, Ciardiello F, Di Mauro A, Mignogna C, Ferrara G, Budillon A, Megchelenbrink WL, Del Gaudio N, Conte M, Merciai F, Campiglia P, Altucci L, Carafa V, Sommella E, Benedetti R. Inverse FASN and LDHA correlation drives metabolic resistance in breast cancer. J Transl Med 2024; 22:676. [PMID: 39044184 PMCID: PMC11267768 DOI: 10.1186/s12967-024-05517-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Breast cancer manifests as a heterogeneous pathology marked by complex metabolic reprogramming essential to satisfy its energy demands. Oncogenic signals boost the metabolism, modifying fatty acid synthesis and glucose use from the onset to progression and therapy resistant-forms. However, the exact contribution of metabolic dependencies during tumor evolution remains unclear. METHODS In this study, we elucidate the connection between FASN and LDHA, pivotal metabolic genes, and their correlation with tumor grade and therapy response using datasets from public repositories. Subsequently, we evaluated the metabolic and proliferative functions upon FASN and LDHA inhibition in breast cancer models. Lastly, we integrated metabolomic and lipidomic analysis to define the contributions of metabolites, lipids, and precursors to the metabolic phenotypes. RESULTS Collectively, our findings indicate metabolic shifts during breast cancer progression, unvealling two distinct functional energy phenotypes associated with aggressiveness and therapy response. Specifically, FASN exhibits reduced expression in advance-grade tumors and therapy-resistant forms, whereas LDHA demonstrates higher expression. Additionally, the biological and metabolic impact of blocking the enzymatic activity of FASN and LDHA was correlated with resistant conditions. CONCLUSIONS These observations emphasize the intrinsic metabolic heterogeneity within breast cancer, thereby highlighting the relevance of metabolic interventions in the field of precision medicine.
Collapse
Affiliation(s)
- Chiara Papulino
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Ugo Chianese
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Ahmad Ali
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Gregorio Favale
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Concetta Tuccillo
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Annabella Di Mauro
- Pathology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Chiara Mignogna
- Pathology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Gerardo Ferrara
- Pathology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Alfredo Budillon
- Scientific Directorate, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | | | - Nunzio Del Gaudio
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Mariarosaria Conte
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Fabrizio Merciai
- Department of Pharmacy (DIFARMA), University of Salerno, 84084, Salerno, Italy
| | - Pietro Campiglia
- Department of Pharmacy (DIFARMA), University of Salerno, 84084, Salerno, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
- Biogem Institute of Molecular and Genetic Biology, 83031, Ariano Irpino, Italy
- Institute of Endocrinology and Oncology "Gaetano Salvatore" (IEOS), 80131, Naples, Italy
- Program of Medical Epigenetics, Vanvitelli Hospital, Naples, Italy
| | - Vincenzo Carafa
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy.
- Biogem Institute of Molecular and Genetic Biology, 83031, Ariano Irpino, Italy.
| | - Eduardo Sommella
- Department of Pharmacy (DIFARMA), University of Salerno, 84084, Salerno, Italy.
| | - Rosaria Benedetti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy.
- Program of Medical Epigenetics, Vanvitelli Hospital, Naples, Italy.
| |
Collapse
|
5
|
Cigliano A, Simile MM, Vidili G, Pes GM, Dore MP, Urigo F, Cossu E, Che L, Feo C, Steinmann SM, Ribback S, Pascale RM, Evert M, Chen X, Calvisi DF. Fatty Acid Synthase Promotes Hepatocellular Carcinoma Growth via S-Phase Kinase-Associated Protein 2/p27 KIP1 Regulation. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1160. [PMID: 39064589 PMCID: PMC11278665 DOI: 10.3390/medicina60071160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/05/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: Aberrant upregulation of fatty acid synthase (FASN), catalyzing de novo synthesis of fatty acids, occurs in various tumor types, including human hepatocellular carcinoma (HCC). Although FASN oncogenic activity seems to reside in its pro-lipogenic function, cumulating evidence suggests that FASN's tumor-supporting role might also be metabolic-independent. Materials and Methods: In the present study, we show that FASN inactivation by specific small interfering RNA (siRNA) promoted the downregulation of the S-phase kinase associated-protein kinase 2 (SKP2) and the consequent induction of p27KIP1 in HCC cell lines. Results: Expression levels of FASN and SKP2 directly correlated in human HCC specimens and predicted a dismal outcome. In addition, forced overexpression of SKP2 rendered HCC cells resistant to the treatment with the FASN inhibitor C75. Furthermore, FASN deletion was paralleled by SKP2 downregulation and p27KIP1 induction in the AKT-driven HCC preclinical mouse model. Moreover, forced overexpression of an SKP2 dominant negative form or a p27KIP1 non-phosphorylatable (p27KIP1-T187A) construct completely abolished AKT-dependent hepatocarcinogenesis in vitro and in vivo. Conclusions: In conclusion, the present data indicate that SKP2 is a critical downstream effector of FASN and AKT-dependent hepatocarcinogenesis in liver cancer, envisaging the possibility of effectively targeting FASN-positive liver tumors with SKP2 inhibitors or p27KIP1 activators.
Collapse
Affiliation(s)
- Antonio Cigliano
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Maria M. Simile
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Gianpaolo Vidili
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Giovanni M. Pes
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Maria P. Dore
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Francesco Urigo
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Eleonora Cossu
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Fracisco, CA 94143, USA; (L.C.); (X.C.)
| | - Claudio Feo
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Sara M. Steinmann
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, 17489 Greifswald, Germany;
| | - Rosa M. Pascale
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Fracisco, CA 94143, USA; (L.C.); (X.C.)
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Diego F. Calvisi
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| |
Collapse
|
6
|
Serhan HA, Bao L, Cheng X, Qin Z, Liu CJ, Heth JA, Udager AM, Soellner MB, Merajver SD, Morikawa A, Merrill NM. Targeting fatty acid synthase in preclinical models of TNBC brain metastases synergizes with SN-38 and impairs invasion. NPJ Breast Cancer 2024; 10:43. [PMID: 38858374 PMCID: PMC11164988 DOI: 10.1038/s41523-024-00656-0] [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: 01/30/2024] [Accepted: 06/01/2024] [Indexed: 06/12/2024] Open
Abstract
Fatty acid synthesis (FAS) has been shown to play a key role in the survival of brain-metastatic (BM) breast cancer. We demonstrate that the fatty acid synthase inhibitor TVB-2640 synergizes with the topoisomerase inhibitor SN-38 in triple-negative breast cancer (TNBC) BM cell lines, upregulates FAS and downregulates cell cycle progression gene expression, and slows the motility of TNBC BM cell lines. The combination of SN-38 and TVB-2640 warrants further consideration as a potential therapeutic option in TNBC BMs.
Collapse
Affiliation(s)
- Habib A Serhan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Liwei Bao
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xu Cheng
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Zhaoping Qin
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chia-Jen Liu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jason A Heth
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Aaron M Udager
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Matthew B Soellner
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sofia D Merajver
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Aki Morikawa
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nathan M Merrill
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
7
|
Sun L, Tuo Z, Chen X, Wang H, Lyu Z, Li G. Identification of cell differentiation trajectory-related gene signature to reveal the prognostic significance and immune landscape in prostate cancer based on multiomics analysis. Heliyon 2024; 10:e27628. [PMID: 38510027 PMCID: PMC10950568 DOI: 10.1016/j.heliyon.2024.e27628] [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: 01/02/2024] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024] Open
Abstract
Background In the context of prostate cancer (PCa), the occurrence of biochemical recurrence (BCR) stands out as a pivotal factor significantly impacting prognosis, potentially leading to metastasis and mortality. However, the early detection of BCR poses a substantial challenge for PCa patients. There is an urgent need to pinpoint hub genes that can serve as predictive indicators for BCR in PCa patients. Methods Our primary goal was to identify cell differentiation trajectory-related gene signature in PCa patients by pseudo-time trajectory analysis. We further explored the functional enrichment of overlapped marker genes and probed clinically relevant modules and BCR-related genes using Weighted Gene Co-expression Network Analysis (WGCNA) in PCa patients. Key genes predicting recurrence-free survival were meticulously identified through univariate and multivariate Cox regression analyses. Subsequently, these genes were utilized to construct a prognostic gene signature, the expression, predictive efficacy, putative functions, and immunological landscape of which were thoroughly validated. Additionally, we employed immunohistochemistry (IHC) and a western blotting assay to quantify the expression of PYCR1 in clinical samples. Results Our single-cell RNA (scRNA) sequencing analysis unveiled three subgroups characterized by distinct differentiation trajectories, and the marker genes associated with these groups were extracted from PCa patients. These marker genes successfully classified the PCa sample into two molecular subtypes, demonstrating a robust correlation with clinical characteristics and recurrence-free survival. Through WGCNA and Lasso analysis, we identified four hub genes (KLK3, CD38, FASN, and PYCR1) to construct a risk profile of prognostic genes linked to BCR. Notably, the high-risk patient group exhibited elevated levels of B cell naive, Macrophage M0, and Macrophage M2 infiltration, while the low-risk group displayed higher levels of T cells CD4 memory activated and monocyte infiltration. Furthermore, IHC and western blotting assays confirmed the heightened expression of PYCR1 in PCa tissues. Conclusion This study leveraged the differentiation trajectory and genetic variability of the microenvironment to uncover crucial prognostic genes associated with BCR in PCa patients. These findings present novel perspectives for tailoring treatment strategies for PCa patients on an individualized basis.
Collapse
Affiliation(s)
- Liangxue Sun
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, China
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xin Chen
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Huming Wang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Zhaojie Lyu
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Guangyuan Li
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, China
- The Lu’ an Hospital Affiliated to Anhui Medical University, Lu’ an, China
- The Lu’ an People’s Hospital, Lu’ an, China
| |
Collapse
|
8
|
Bandi DSR, Sarvesh S, Farran B, Nagaraju GP, El-Rayes BF. Targeting the metabolism and immune system in pancreatic ductal adenocarcinoma: Insights and future directions. Cytokine Growth Factor Rev 2023; 71-72:26-39. [PMID: 37407355 DOI: 10.1016/j.cytogfr.2023.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/07/2023]
Abstract
Pancreatic cancer, specifically pancreatic ductal adenocarcinoma (PDAC), presents a challenging landscape due to its complex nature and the highly immunosuppressive tumor microenvironment (TME). This immunosuppression severely limits the effectiveness of immune-based therapies. Studies have revealed the critical role of immunometabolism in shaping the TME and influencing PDAC progression. Genetic alterations, lysosomal dysfunction, gut microbiome dysbiosis, and altered metabolic pathways have been shown to modulate immunometabolism in PDAC. These metabolic alterations can significantly impact immune cell functions, including T-cells, myeloid-derived suppressor cells (MDSCs), and macrophages, evading anti-tumor immunity. Advances in immunotherapy offer promising avenues for overcoming immunosuppressive TME and enhancing patient outcomes. This review highlights the challenges and opportunities for future research in this evolving field. By exploring the connections between immunometabolism, genetic alterations, and the microbiome in PDAC, it is possible to tailor novel approaches capable of improving immunotherapy outcomes and addressing the limitations posed by immunosuppressive TME. Ultimately, these insights may pave the way for improved treatment options and better outcomes for PDAC patients.
Collapse
Affiliation(s)
- Dhana Sekhar Reddy Bandi
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA
| | - Sujith Sarvesh
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA
| | - Batoul Farran
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA.
| | - Bassel F El-Rayes
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA.
| |
Collapse
|