1
|
Poswal J, Mandal CC. Lipid metabolism dysregulation for bone metastasis and its prevention. Expert Rev Anticancer Ther 2025:1-17. [PMID: 40219980 DOI: 10.1080/14737140.2025.2492784] [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: 02/16/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
INTRODUCTION Bone metastasis often develops in advanced malignancies. Lipid metabolic dysregulation might play pivotal role in cancer progression and subsequent deterioration of bone health at metastatic condition. In-depth understanding of lipid reprogramming in metastasized cancer cells and other stromal cells including bone marrow adipocyte (BMA) is an urgent need to develop effective therapy. AREA COVERED This paper emphasizes providing an overview of multifaceted role of dysregulated lipids and BMA in cancer cells in association with bone metastasis by utilizing search terms lipid metabolism, lipid and metastasis in PubMed. This study extends to address mechanism linked with lipid metabolism and various crucial genes (e.g. CSF-1, RANKL, NFkB and NFATc1) involved in bone metastasis. This review examines therapeutic strategies targeting lipid metabolism to offer potential avenues to disrupt lipid-driven metastasis. EXPERT OPINION On metastatic condition, dysregulated lipid molecules especially in BMA and other stromal cells not only favors cancer progression but also potentiate lipid reprogramming within cancer cells. Distinct dysregulated lipid-metabolism associated genes may act as biomarker, and targeting these is challenging task for specific treatment. Curbing function of bone resorption associated genes by lipid controlling drugs (e.g. statins, omega-3 FA and metformin) may provide additional support to curtail lipid-associated bone metastasis.
Collapse
Affiliation(s)
- Jyoti Poswal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| |
Collapse
|
2
|
Bhat M, Saha P, Narasimhan M, Shelar A, Hole A, Murali Krishna C, Govekar R. Analysis of lipids by Raman spectroscopy and mass spectrometry provides a detection tool and mechanistic insight into imatinib resistance in CML-BC. Biochim Biophys Acta Gen Subj 2025; 1869:130771. [PMID: 39938699 DOI: 10.1016/j.bbagen.2025.130771] [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: 09/11/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
Resistance to tyrosine kinase inhibitors (TKIs) is a major challenge in the treatment of chronic myeloid leukemia (CML). Established tests based on the known mechanisms of resistance in the initial chronic phase (CP) confirm resistance, reveal the underlying reason and thereby direct treatment modifications. In the terminal phase of blast crisis (BC), however, additional partially identified mechanisms of resistance exist which necessitates developing modalities to detect resistance regardless of the underlying mechanism and concurrent exploration of the resistance mechanism to assist in identification of appropriate drug targets. In this study both the clinical objectives were achieved by analysing lipids in BC cells, sensitive and resistant to TKIs, using the complementary strengths of distinct analytical technologies. Raman spectroscopy, through the spectral signatures with lipids as a significant differentiating component could segregate resistant from sensitive cells in the Principal Component Analysis (PCA) and Principal Component based Linear Discriminant Analysis (PC-LDA). This provided a tool to rapidly detect resistance in CML-BC despite unclear mechanism of TKI resistance. The depth of coverage achievable by mass spectrometry allowed the generation of quantitative differential profile of individual lipids in resistant cells. The alterations were in diverse classes of lipids which are involved in cell signalling and inhibition studies could link these alterations to modulation of phospholipase A2 (PLA2) levels mediated by p38 mitogen activated protein kinase (p38MAPK), which is causally associated with TKI resistance in CML-BC. Together, lipid analysis using the two platforms, contributed to the detection and mechanistic understanding of imatinib resistance in CML-BC.
Collapse
Affiliation(s)
- Manish Bhat
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai 400094, India
| | - Panchali Saha
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai 400094, India
| | - Mythreyi Narasimhan
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ashutosh Shelar
- Shimadzu Analytical (India) Pvt. Ltd., Mumbai 400 059, India
| | - Arti Hole
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India
| | - C Murali Krishna
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai 400094, India.
| | - Rukmini Govekar
- Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai 410210, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai 400094, India.
| |
Collapse
|
3
|
Hossain MK, Unger L, Larsen U, Altankhuyag A, Legøy TA, Paulo JA, Vethe H, Ghila L. Mapping the initial effects of carcinogen-induced oncogenic transformation in the mouse bladder. Exp Cell Res 2025; 446:114452. [PMID: 39988124 DOI: 10.1016/j.yexcr.2025.114452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
Characterizing the initial stages of oncogenic transformation allows the identification of tumor-promoting processes before the inherent clonal selection of the aggressive clones. Here, we used global proteomics, genetic cell tracing, and immunofluorescence to dynamically map the very early stages of cancer initiation in a mouse model of bladder cancer. We observed a very rapid and incremental proteome dysregulation, with changes in the energy metabolism, proliferation and immune signatures dominating the landscape. The changes in the lipid metabolism were immediate and defined by an increase fatty acid metabolism and lipid transport, followed by the activation of the immune landscape. Alongside the changes in the immune signature and lipid metabolism, we also mapped a clear increase in the cell cycle-related pathways and proliferation. Proliferation was mainly restricted to the basal epithelial layer rapidly leading to urothelium thickening, despite the progressive loss of the superficial layer. Moreover, we observed a tilt in the energy balance towards increased glucose metabolism, probably characterizing cells of the tumor microenvironment. All of the observed proteome signature changes were persistent, being retained and sometimes intensified or diversified along the timeline. The signatures observed in this pilot suggest these processes as potentially targetable drivers of the future neoplastic transformations in the bladder.
Collapse
Affiliation(s)
- Md Kaykobad Hossain
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | - Lucas Unger
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | - Ulrik Larsen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | | | - Thomas Aga Legøy
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Heidrun Vethe
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Norway
| | - Luiza Ghila
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway.
| |
Collapse
|
4
|
Dakal TC, Xiao F, Bhusal CK, Sabapathy PC, Segal R, Chen J, Bai X. Lipids dysregulation in diseases: core concepts, targets and treatment strategies. Lipids Health Dis 2025; 24:61. [PMID: 39984909 PMCID: PMC11843775 DOI: 10.1186/s12944-024-02425-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/30/2024] [Indexed: 02/23/2025] Open
Abstract
Lipid metabolism is a well-regulated process essential for maintaining cellular functions and energy homeostasis. Dysregulation of lipid metabolism is associated with various conditions, including cardiovascular diseases, neurodegenerative disorders, and metabolic syndromes. This review explores the mechanisms underlying lipid metabolism, emphasizing the roles of key lipid species such as triglycerides, phospholipids, sphingolipids, and sterols in cellular physiology and pathophysiology. It also examines the genetic and environmental factors contributing to lipid dysregulation and the challenges of diagnosing and managing lipid-related disorders. Recent advancements in lipid-lowering therapies, including PCSK9 inhibitors, ezetimibe, bempedoic acid, and olpasiran, provide promising treatment options. However, these advancements are accompanied by challenges related to cost, accessibility, and patient adherence. The review highlights the need for personalized medicine approaches to address the interplay between genetics and environmental factors in lipid metabolism. As lipidomics and advanced diagnostic tools continue to progress, a deeper understanding of lipid-related disorders could pave the way for more effective therapeutic strategies.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Mohanlal Sukhadia, University, Udaipur, 313001, India
| | - Feng Xiao
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China
| | - Chandra Kanta Bhusal
- Aarupadai Veedu Medical College and Hospital, VMRF-DU, Pondicherry, 607402, India
- Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | | | - Rakesh Segal
- Aarupadai Veedu Medical College and Hospital, VMRF-DU, Pondicherry, 607402, India
- Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Juan Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, China.
| | - Xiaodong Bai
- Department of Plastic Surgery, Southern University of Science and Technology Hospital, Shenzhen, 518055, China.
| |
Collapse
|
5
|
Aden D, Sureka N, Zaheer S, Chaurasia JK, Zaheer S. Metabolic Reprogramming in Cancer: Implications for Immunosuppressive Microenvironment. Immunology 2025; 174:30-72. [PMID: 39462179 DOI: 10.1111/imm.13871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Cancer is a complex and heterogeneous disease characterised by uncontrolled cell growth and proliferation. One hallmark of cancer cells is their ability to undergo metabolic reprogramming, which allows them to sustain their rapid growth and survival. This metabolic reprogramming creates an immunosuppressive microenvironment that facilitates tumour progression and evasion of the immune system. In this article, we review the mechanisms underlying metabolic reprogramming in cancer cells and discuss how these metabolic alterations contribute to the establishment of an immunosuppressive microenvironment. We also explore potential therapeutic strategies targeting metabolic vulnerabilities in cancer cells to enhance immune-mediated anti-tumour responses. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02044861, NCT03163667, NCT04265534, NCT02071927, NCT02903914, NCT03314935, NCT03361228, NCT03048500, NCT03311308, NCT03800602, NCT04414540, NCT02771626, NCT03994744, NCT03229278, NCT04899921.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
6
|
Singer M, Hamdy R, Elsayed TM, Husseiny MI. The Mechanisms and Therapeutic Implications of Metabolic Communication in the Tumor-Immune Microenvironment. METABOLIC DYNAMICS IN HOST-MICROBE INTERACTION 2025:291-315. [DOI: 10.1007/978-981-96-1305-2_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
|
7
|
Guo X, Wang F, Li X, Luo Q, Liu B, Yuan J. Mitochondrial cholesterol metabolism related gene model predicts prognosis and treatment response in hepatocellular carcinoma. Transl Cancer Res 2024; 13:6623-6644. [PMID: 39816559 PMCID: PMC11730194 DOI: 10.21037/tcr-24-1153] [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: 07/06/2024] [Accepted: 10/25/2024] [Indexed: 01/18/2025]
Abstract
Background The persistently high mortality and morbidity rates of hepatocellular carcinoma (HCC) remain a global concern. Notably, the disruptions in mitochondrial cholesterol metabolism (MCM) play a pivotal role in the progression and development of HCC, underscoring the significance of this metabolic pathway in the disease's etiology. The purpose of this research was to investigate genes associated with MCM and develop a model for predicting the prognostic features of patients with HCC. Methods MCM-related genes (MCMGs) were identified through The Cancer Genome Atlas (TCGA), The Molecular Signatures Database (MsigDB), and the Mitocarta3.0 databases. Differential gene expression analysis and least absolute shrinkage and selection operator (LASSO) Cox regression analysis were performed using R software to construct a MCM-related model. This model underwent further analysis for somatic mutations, single sample gene set enrichment analysis (ssGSEA), stromal and immune cell estimation, immune checkpoint evaluation, and drug susceptibility prediction to assess the tumor microenvironment (TME) and therapeutic responses. The mRNA expression levels of the genes associated with the model were quantified using real-time fluorescence quantitative polymerase chain reaction (RT-qPCR). Results The model, which included six MCMGs (ACADL, ACLY, TXNRD1, DTYMK, ACAT1, and FLAD1), divided all patients (age ≤65 vs. >65 years, P<0.001; male vs. female, ns) into a high-risk group and a low-risk group. The high-risk group showed a higher mortality rate and lower survival rate with AUC of 0.785, 0.752, 0.756, 0.774 and 0.759 for the 1-, 2-, 3-, 4-, and 5-year respectively. A nomogram based on risk score, stage, T, and M had a better prognostic accuracy, with AUC of 0.808, 0.796, 0.811, 0.824 and 0.795 for the 1-, 2-, 3-, 4-, and 5-year respectively. The high-risk group showed enrichment in cell cycle, cell division, and chromosome processes, and a significantly higher tumor mutation burden (TMB) value compared to the low-risk group. Further immune infiltration analysis indicated a significantly reduction in the abundances of some immune cells (activated CD4 T cells, type 2 helper T cells, and neutrophils) and significantly higher expression levels of some immune checkpoint (CD80, CTLA4, HAVCR2, and TNFRSF4) in the high-risk group. Moreover, the risk score was associated with the response to immune checkpoint inhibitors (ICIs) therapy and efficiencies of multiple chemotherapy drugs. Conclusions This study developed a prognostic model based on MCMGs, which can predict the prognosis of liver cancer patients and their response to immunotherapy and chemotherapy. The model may provide new strategies to enhance the prognosis and treatment of HCC.
Collapse
Affiliation(s)
- Xuna Guo
- Department of Biomedical Engineering, School of Life Sciences, Guangxi Medical University, Nanning, China
| | - Feng Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Xuejing Li
- Department of Physiology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Qiuqian Luo
- Department of Physiology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Bihan Liu
- Department of Biomedical Engineering, School of Life Sciences, Guangxi Medical University, Nanning, China
| | - Jianhui Yuan
- Department of Biomedical Engineering, School of Life Sciences, Guangxi Medical University, Nanning, China
- Department of Physiology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
- Department of Physics, Guangxi Medical University, Nanning, China
| |
Collapse
|
8
|
Ciołczyk-Wierzbicka D, Zarzycka M, Placha W, Zemanek G, Wierzbicki K. Treatment of Melanoma Cells with Chloroquine and Everolimus Activates the Apoptosis Process and Alters Lipid Redistribution. Int J Mol Sci 2024; 25:12278. [PMID: 39596342 PMCID: PMC11594807 DOI: 10.3390/ijms252212278] [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: 09/22/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
The balance between apoptosis and autophagy plays a key role in cancer biology and treatment strategies. The aim of this study was to assess the effect of the mTOR kinase inhibitor everolimus and chloroquine on the regulation of proliferation, caspase-3 activation, and apoptosis in melanoma cells. We studied the activity of caspase-3 and the levels of caspase-3 and -9 using the Western blot technique. Cellular apoptosis was examined using a DNA fragmentation assay, and changes in the cell nucleus and cytoskeleton were examined using fluorescence microscopy DAPI, OA/IP. We also studied the rearrangement of lipid structures using fluorescent dyes: Nile Red and Nile Blue. A low nanomolar concentration of the mTOR kinase inhibitor everolimus in combination with chloroquine activated the apoptosis process and decreased cell proliferation. These changes were accompanied by an obvious change in cell morphology and rearrangement of lipid structures. Alterations in lipid redistribution accompanying the process of apoptosis and autophagy are among the first to occur in the cell and can be easily monitored in in vitro studies. The combination of mTOR inhibitors and chloroquine represents a promising area of research in cancer therapy. It has the potential to enhance treatment efficacy through complementary mechanisms.
Collapse
Affiliation(s)
- Dorota Ciołczyk-Wierzbicka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul. Kopernika 7, 31-034 Cracow, Poland; (M.Z.); (W.P.); (G.Z.)
| | - Marta Zarzycka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul. Kopernika 7, 31-034 Cracow, Poland; (M.Z.); (W.P.); (G.Z.)
| | - Wojciech Placha
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul. Kopernika 7, 31-034 Cracow, Poland; (M.Z.); (W.P.); (G.Z.)
| | - Grzegorz Zemanek
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul. Kopernika 7, 31-034 Cracow, Poland; (M.Z.); (W.P.); (G.Z.)
| | - Karol Wierzbicki
- Department of Cardiovascular Surgery and Transplantology, Institute of Cardiology, Jagiellonian University, John Paul II Hospital, ul. Prądnicka 80, 31-202 Cracow, Poland
| |
Collapse
|
9
|
Tanaka H, Fukawa Y, Yamamoto K, Tanimoto K, Takemoto A, Mori T, Hasumi H, Kinoshita M, Kanazawa T, Furukawa A, Kimura K, Sato H, Hirakawa A, Fukuda S, Waseda Y, Yoshida S, Campbell SC, Fujii Y. Prognostic Impact and Genomic Backgrounds of Renal Parenchymal Infiltration or Micronodular Spread in Nonmetastatic Clear Cell Renal Cell Carcinoma. Mod Pathol 2024; 37:100590. [PMID: 39142537 DOI: 10.1016/j.modpat.2024.100590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 06/28/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024]
Abstract
A subset of clear cell renal cell carcinomas (ccRCCs) exhibits various growth patterns that infiltrate the normal renal parenchyma; however, our understanding of its association with cancer aggressiveness is incomplete. Here, we show that the morphology of the tumor interface with normal renal parenchyma is robustly associated with cancer recurrence after surgery, even when compared with the TNM staging system or the World Health Organization/International Society of Urological Pathology (WHO/ISUP) nuclear grade in nonmetastatic ccRCC. Hematoxylin and eosin-stained slides of whole tissue sections from surgical specimens were analyzed using a cohort of 331 patients with nonmetastatic ccRCC treated with radical nephrectomy. The patients were classified into 10 subgroups based on our classification algorithms for assessing the tumor interface with normal renal parenchyma. Among the 10 subgroups, 4 subgroups consisting of 40 patients (12%) were identified to have aggressive forms of nonmetastatic ccRCC associated with poor prognosis and unified as renal parenchymal infiltration or micronodular spread (RPI/MNS) phenotypes. Multivariable analyses showed that RPI/MNS phenotypes were robustly associated with shorter disease-free survival, independently of existing pathological factors including the TNM staging system and WHO/ISUP nuclear grade. The hazard ratio was highest for RPI/MNS (4.62), followed by WHO/ISUP grades 3 to 4 (2.11) and ≥pT3a stage (2.05). In addition, we conducted genomic analyses using next-generation sequencing of infiltrative lesions in 18 patients with RPI/MNS and tumor lesions in 33 patients without RPI/MNS. Results showed that alterations in SETD2 and TSC1 might be associated with RPI/MNS phenotypes, whereas alterations in PBRM1 might be associated with non-RPI/MNS phenotypes. These data suggest that RPI/MNS may be associated with aggressive genomic backgrounds of ccRCC, although more comprehensive analyses with a larger sample size are required. Future studies may further elucidate the clinical implications of RPI/MNS, particularly for deciding the indication of adjuvant treatment after nephrectomy.
Collapse
Affiliation(s)
- Hajime Tanaka
- Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Yuki Fukawa
- Department of Pathology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kouhei Yamamoto
- Department of Pathology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kousuke Tanimoto
- Research Core, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akira Takemoto
- Bioresource Research Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takayasu Mori
- Department of Nephrology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hisashi Hasumi
- Department of Urology, Yokohama City University, Yokohama, Japan
| | - Mayumi Kinoshita
- Department of Pathology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Clinical Laboratory Medicine, Faculty of Health Science Technology, Bunkyo Gakuin University, Tokyo, Japan
| | - Takumi Kanazawa
- Department of Pathology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Clinical Laboratory Medicine, Faculty of Health Science Technology, Bunkyo Gakuin University, Tokyo, Japan
| | - Asuka Furukawa
- Department of Pathology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koichiro Kimura
- Department of Radiology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Sato
- Department of Clinical Biostatistics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shohei Fukuda
- Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuma Waseda
- Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Soichiro Yoshida
- Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Steven C Campbell
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio
| | - Yasuhisa Fujii
- Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
10
|
Singh MK, Han S, Kim S, Kang I. Targeting Lipid Metabolism in Cancer Stem Cells for Anticancer Treatment. Int J Mol Sci 2024; 25:11185. [PMID: 39456967 PMCID: PMC11508222 DOI: 10.3390/ijms252011185] [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: 09/06/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Cancer stem cells (CSCs), or tumor-initiating cells (TICs), are small subpopulations (0.0001-0.1%) of cancer cells that are crucial for cancer relapse and therapy resistance. The elimination of each CSC is essential for achieving long-term remission. Metabolic reprogramming, particularly lipids, has a significant impact on drug efficacy by influencing drug diffusion, altering membrane permeability, modifying mitochondrial function, and adjusting the lipid composition within CSCs. These changes contribute to the development of chemoresistance in various cancers. The intricate relationship between lipid metabolism and drug resistance in CSCs is an emerging area of research, as different lipid species play essential roles in multiple stages of autophagy. However, the link between autophagy and lipid metabolism in the context of CSC regulation remains unclear. Understanding the interplay between autophagy and lipid reprogramming in CSCs could lead to the development of new approaches for enhancing therapies and reducing tumorigenicity in these cells. In this review, we explore the latest findings on lipid metabolism in CSCs, including the role of key regulatory enzymes, inhibitors, and the contribution of autophagy in maintaining lipid homeostasis. These recent findings may provide critical insights for identifying novel pharmacological targets for effective anticancer treatment.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (S.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (S.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sungsoo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (S.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (S.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
11
|
Paunovic M, Stojanovic A, Pokimica B, Martacic JD, Cvetkovic Z, Ivanovic N, Vucic V. Metabolic Reprogramming of Phospholipid Fatty Acids as a Signature of Lung Cancer Type. Cancers (Basel) 2024; 16:3320. [PMID: 39409945 PMCID: PMC11475191 DOI: 10.3390/cancers16193320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Lung cancer is one of the leading causes of cancer-related mortality. Non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) differ in aggressiveness, proliferation speed, metastasis propensity, and prognosis. Since tumor cells notably change lipid metabolism, especially phospholipids and fatty acids (FA), this study aimed to identify FA alterations in lung cancer tissues. Methods: Our study included patients with newly diagnosed, histologically confirmed SCLC (n = 27) and NSCLC (n = 37). Samples were collected from both malignant and healthy tissues from each patient, providing they were within subject design. Results: In both NSCLC and SCLC tumor tissues, FA contents were shifted toward pro-inflammatory profiles, with increased levels of some individual n-6 polyunsaturated FA (PUFA), particularly arachidonic acid, and elevated activity of Δ6 desaturase. Compared to healthy counterparts, lower levels of alpha-linolenic acid (18:3n-3) and total saturated FA (SFA) were found in NSCLC, while decreased levels of linoleic acid (18:2n-6) and all individual n-3 FA were found in SCLC tissue in comparison to the healthy tissue control. When mutually compared, SCLC tissue had higher levels of total SFA, especially stearic acid, while higher levels of linoleic acid, total PUFA, and n-3 and n-6 PUFA were detected in NSCLC. Estimated activities of Δ6 desaturase and elongase were higher in SCLC than in NSCLC. Conclusions: Our findings indicate a notable impairment of lipid metabolism in two types of lung cancer tissues. These type-specific alterations may be associated with differences in their progression and also point out different therapeutic targets.
Collapse
Affiliation(s)
- Marija Paunovic
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (B.P.); (J.D.M.)
| | - Ana Stojanovic
- Department of Pulmonology, University Hospital Medical Center (UHMC) “Bezanijska kosa”, 11000 Belgrade, Serbia;
| | - Biljana Pokimica
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (B.P.); (J.D.M.)
| | - Jasmina Debeljak Martacic
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (B.P.); (J.D.M.)
| | - Zorica Cvetkovic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (Z.C.); (N.I.)
- Department of Hematology, University Hospital Medical Center Zemun, 11080 Belgrade, Serbia
| | - Nebojsa Ivanovic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (Z.C.); (N.I.)
- Department of Surgical Oncology, University Hospital Medical Center (UHMC) “Bezanijska kosa”, 11000 Belgrade, Serbia
| | - Vesna Vucic
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (B.P.); (J.D.M.)
| |
Collapse
|
12
|
Kosałka-Węgiel J, Pacholczak-Madej R, Dziedzic R, Siwiec-Koźlik A, Spałkowska M, Milewski M, Zaręba L, Bazan-Socha S, Korkosz M. Malignancy in systemic lupus erythematosus: relation to disease characteristics in 92 patients - a single center retrospective study. Rheumatol Int 2024; 44:1701-1713. [PMID: 38850326 PMCID: PMC11343918 DOI: 10.1007/s00296-024-05623-3] [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: 05/07/2024] [Accepted: 05/25/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with a variable clinical manifestation, potentially leading to death. Importantly, patients with SLE have an increased risk of neoplastic disorders. Thus, this study aimed to comprehensively evaluate the clinical and laboratory characteristics of patients with SLE and with or without malignancy. METHODS We conducted a retrospective analysis of medical records of 932 adult Caucasian patients with SLE treated at the University Hospital in Kraków, Poland, from 2012 to 2022. We collected demographic, clinical, and laboratory characteristics, but also treatment modalities with disease outcomes. RESULTS Among 932 patients with SLE, malignancy was documented in 92 (9.87%), with 7 (7.61%) patients experiencing more than one such complication. Non-hematologic malignancies were more prevalent (n = 77, 83.7%) than hematologic malignancies (n = 15, 16.3%). Patients with SLE and malignancy had a higher mean age of SLE onset and a longer mean disease duration than patients without malignancy (p < 0.001 and p = 0.027, respectively). The former group also presented more frequently with weight loss (odds ratio [OR] = 2.62, 95% confidence interval [CI] 1.61-4.23, p < 0.001), fatigue/weakness (OR = 2.10, 95% CI 1.22-3.77, p = 0.005), and fever (OR = 1.68, 95% CI 1.06-2.69, p = 0.024). In the malignancy-associated group, we noticed a higher prevalence of some clinical manifestations, such as pulmonary hypertension (OR = 3.47, 95% CI 1.30-8.42, p = 0.007), lung involvement (OR = 2.64, 95% CI 1.35-4.92, p = 0.003) with pleural effusion (OR = 2.39, 95% CI 1.43-3.94, p < 0.001), and anemia (OR = 2.24, 95% CI 1.29-4.38, p = 0.006). Moreover, the patients with SLE and malignancy more frequently had internal comorbidities, including peripheral arterial obliterans disease (OR = 3.89, 95% CI 1.86-7.75, p < 0.001), myocardial infarction (OR = 3.08, 95% CI 1.41-6.30, p = 0.003), heart failure (OR = 2.94, 95% CI 1.30-6.17, p = 0.005), diabetes mellitus (OR = 2.15, 95% CI 1.14-3.91, p = 0.011), hypothyroidism (OR = 2.08, 95% CI 1.29-3.34, p = 0.002), arterial hypertension (OR = 1.97, 95% CI 1.23-3.23, p = 0.003), and hypercholesterolemia (OR = 1.87, 95% CI 1.18-3.00, p = 0.006). Patients with SLE and malignancy were treated more often with aggressive immunosuppressive therapies, including cyclophosphamide (OR = 2.07, 95% CI 1.30-3.28, p = 0.002), however median cumulative cyclophosphamide dose in malignancy-associated SLE subgroup was 0 g (0-2 g). Interestingly, over a median follow-up period of 14 years (ranges: 8-22 years) a total of 47 patients with SLE died, with 16 cases (5.28%) in the malignancy-associated SLE group and 31 cases (5.73%) in the non-malignancy SLE group (p = 0.76). The most common causes of death were infections (21.28%) and SLE exacerbation (8.51%). CONCLUSION The study highlights the relatively frequent presence of malignancies in patients with SLE, a phenomenon that demands oncological vigilance, especially in patients with a severe clinical course and comorbidities, to improve long-term outcomes in these patients.
Collapse
Affiliation(s)
- Joanna Kosałka-Węgiel
- Department of Rheumatology and Immunology, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-688, Poland.
- Department of Rheumatology, Immunology and Internal Medicine, University Hospital, Jakubowskiego 2, Kraków, 30-688, Poland.
| | - Renata Pacholczak-Madej
- Department of Gynaecological Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, Kraków, 31-115, Poland
- Department of Chemotherapy, The District Hospital, Szpitalna 22, Sucha Beskidzka, 34-200, Poland
- Department of Anatomy, Jagiellonian University Medical College, Kopernika 12, Kraków, 31-034, Poland
| | - Radosław Dziedzic
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Św. Łazarza 16, Kraków, 31-530, Poland
| | - Andżelika Siwiec-Koźlik
- Department of Rheumatology, Immunology and Internal Medicine, University Hospital, Jakubowskiego 2, Kraków, 30-688, Poland
| | - Magdalena Spałkowska
- Department of Dermatology, Jagiellonian University Medical College, Botaniczna 3, Kraków, 31-501, Poland
| | - Mamert Milewski
- Department of Rheumatology, Immunology and Internal Medicine, University Hospital, Jakubowskiego 2, Kraków, 30-688, Poland
| | - Lech Zaręba
- College of Natural Sciences, Institute of Computer Science, University of Rzeszów, Pigonia 1, Rzeszów, 35-310, Poland
| | - Stanisława Bazan-Socha
- Department of Rheumatology, Immunology and Internal Medicine, University Hospital, Jakubowskiego 2, Kraków, 30-688, Poland
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-688, Poland
| | - Mariusz Korkosz
- Department of Rheumatology and Immunology, Jagiellonian University Medical College, Jakubowskiego 2, Kraków, 30-688, Poland
- Department of Rheumatology, Immunology and Internal Medicine, University Hospital, Jakubowskiego 2, Kraków, 30-688, Poland
| |
Collapse
|
13
|
Cheung SM, Chan KS, Senn N, Husain E, Sharma R, McGoldrick T, Gagliardi T, Masannat Y, He J. Peri-Tumoural Lipid Composition and Hypoxia for Early Immune Response to Neoadjuvant Chemotherapy in Breast Cancer. Int J Mol Sci 2024; 25:9303. [PMID: 39273252 PMCID: PMC11395063 DOI: 10.3390/ijms25179303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
The deregulation of monounsaturated, polyunsaturated, and saturated fatty acids (MUFAs, PUFAs, SFAs) from de novo synthesis and hypoxia are central metabolic features of breast tumour. Early response markers for neoadjuvant chemotherapy (NACT) are critical for stratified treatment for patients with breast cancer, and restoration of lipid metabolism and normoxia might precede observable structural change. In this study, we hypothesised that peri-tumoural lipid composition and hypoxia might be predictive and early response markers in patients with breast cancer undergoing NACT. Female patients with breast cancer were scanned on a 3T clinical MRI scanner at baseline and Cycle1, with acquisition of lipid composition maps of MUFAs, PUFAs, and SFAs, and hypoxia maps of effective transverse relaxation rate R2*. The percentage change in lipid composition and hypoxia at Cycle1 was calculated with reference to baseline. Tumour-associated macrophages were analysed based on immunostaining of CD163 from biopsy and resection, with the percentage change in the resected tumour calculated across the entire NACT. We found no significant difference in lipid composition and R2* between good and poor responders at baseline and Cycle1; however, the correlation between the percentage change in MUFAs and PUFAs against CD163 suggested the modulation in lipids with altered immune response might support the development of targeted therapies.
Collapse
Affiliation(s)
- Sai Man Cheung
- Newcastle Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Kwok-Shing Chan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Nicholas Senn
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Ehab Husain
- Department of Pathology, Aberdeen Royal Infirmary, Aberdeen AB25 2ZN, UK
| | - Ravi Sharma
- Department of Oncology, Aberdeen Royal Infirmary, Aberdeen AB25 2ZN, UK
| | - Trevor McGoldrick
- Department of Oncology, Aberdeen Royal Infirmary, Aberdeen AB25 2ZN, UK
| | - Tanja Gagliardi
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
- Department of Radiology, Royal Marsden Hospital, London SW3 6JJ, UK
| | - Yazan Masannat
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
- Breast Unit, Broomfield Hospital, Mid and South Essex NHS Foundation Trust, Chelmsford CM1 7ET, UK
| | - Jiabao He
- Newcastle Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| |
Collapse
|
14
|
Ahuja S, Sureka N, Zaheer S. Unraveling the intricacies of cancer-associated fibroblasts: a comprehensive review on metabolic reprogramming and tumor microenvironment crosstalk. APMIS 2024. [PMID: 38873945 DOI: 10.1111/apm.13447] [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: 02/02/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are crucial component of tumor microenvironment (TME) which undergo significant phenotypic changes and metabolic reprogramming, profoundly impacting tumor growth. This review delves into CAF plasticity, diverse origins, and the molecular mechanisms driving their continuous activation. Emphasis is placed on the intricate bidirectional crosstalk between CAFs and tumor cells, promoting cancer cell survival, proliferation, invasion, and immune evasion. Metabolic reprogramming, a cancer hallmark, extends beyond cancer cells to CAFs, contributing to the complex metabolic interplay within the TME. The 'reverse Warburg effect' in CAFs mirrors the Warburg effect, involving the export of high-energy substrates to fuel cancer cells, supporting their rapid proliferation. Molecular regulations by key players like p53, Myc, and K-RAS orchestrate this metabolic adaptation. Understanding the metabolic symbiosis between CAFs and tumor cells opens avenues for targeted therapeutic strategies to disrupt this dynamic crosstalk. Unraveling CAF-mediated metabolic reprogramming provides valuable insights for developing novel anticancer therapies. This comprehensive review consolidates current knowledge, shedding light on CAFs' multifaceted roles in the TME and offering potential targets for future therapies.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|