1
|
El-Miligy MMM, Al-Kubeisi AK, Nassra RA, El-Zemity SR, Hazzaa AA. Discovery of new thymol-3,4-disubstituted thiazole hybrids as dual COX-2/5-LOX inhibitors with in vivo proof. J Enzyme Inhib Med Chem 2024; 39:2309171. [PMID: 38291670 PMCID: PMC10833116 DOI: 10.1080/14756366.2024.2309171] [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: 08/23/2023] [Accepted: 01/14/2024] [Indexed: 02/01/2024] Open
Abstract
New thymol-3,4-disubstitutedthiazole hybrids were synthesised as dual COX-2/5-LOX inhibitors. Compounds 6b, 6d, 6e, and 6f displayed in vitro inhibitory activity against COX-2 (IC50= 0.037, 0.042, 0.046, and 0.039 µM) nearly equal to celecoxib (IC50= 0.045 µM). 6b, 6d, and 6f showed SI (379, 341, and 374, respectively) higher than that of celecoxib (327). 6a-l elicited in vitro 5-LOX inhibitory activity higher than quercetin. 6a-f, 6i-l, 7a, and 7c possessed in vivo inhibition of formalin induced paw edoema higher than celecoxib. 6a, 6b, 6f, 6h-l, and 7b showed gastrointestinal safety profile as celecoxib and diclofenac sodium in the population of fasted rats. Induced fit docking and molecular dynamics simulation predicted good fitting of 6b and 6f without changing the packing and globularity of the apo protein. In conclusion, 6b and 6f achieved the target goal as multitarget inhibitors of inflammation.
Collapse
Affiliation(s)
- Mostafa M. M. El-Miligy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | | | - Rasha A. Nassra
- Medical Biochemistry Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Saad R. El-Zemity
- Department of Chemistry and Technology of Pesticides, Faculty of Agriculture, Alexandria University, Alexandria, Egypt
| | - Aly A. Hazzaa
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
2
|
Punyawatthananukool S, Matsuura R, Wongchang T, Katsurada N, Tsuruyama T, Tajima M, Enomoto Y, Kitamura T, Kawashima M, Toi M, Yamanoi K, Hamanishi J, Hisamori S, Obama K, Charoensawan V, Thumkeo D, Narumiya S. Prostaglandin E 2-EP2/EP4 signaling induces immunosuppression in human cancer by impairing bioenergetics and ribosome biogenesis in immune cells. Nat Commun 2024; 15:9464. [PMID: 39487111 PMCID: PMC11530437 DOI: 10.1038/s41467-024-53706-3] [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: 05/30/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
While prostaglandin E2 (PGE2) is produced in human tumor microenvironment (TME), its role therein remains poorly understood. Here, we examine this issue by comparative single-cell RNA sequencing of immune cells infiltrating human cancers and syngeneic tumors in female mice. PGE receptors EP4 and EP2 are expressed in lymphocytes and myeloid cells, and their expression is associated with the downregulation of oxidative phosphorylation (OXPHOS) and MYC targets, glycolysis and ribosomal proteins (RPs). Mechanistically, CD8+ T cells express EP4 and EP2 upon TCR activation, and PGE2 blocks IL-2-STAT5 signaling by downregulating Il2ra, which downregulates c-Myc and PGC-1 to decrease OXPHOS, glycolysis, and RPs, impairing migration, expansion, survival, and antitumor activity. Similarly, EP4 and EP2 are induced upon macrophage activation, and PGE2 downregulates c-Myc and OXPHOS in M1-like macrophages. These results suggest that PGE2-EP4/EP2 signaling impairs both adaptive and innate immunity in TME by hampering bioenergetics and ribosome biogenesis of tumor-infiltrating immune cells.
Collapse
MESH Headings
- Dinoprostone/metabolism
- Humans
- Animals
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Receptors, Prostaglandin E, EP2 Subtype/genetics
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Signal Transduction
- Female
- Tumor Microenvironment/immunology
- Mice
- Energy Metabolism
- Ribosomes/metabolism
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/genetics
- Neoplasms/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Oxidative Phosphorylation
- Glycolysis
- Macrophages/metabolism
- Macrophages/immunology
- Mice, Inbred C57BL
- Cell Line, Tumor
- Immune Tolerance
Collapse
Affiliation(s)
| | - Ryuma Matsuura
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Thamrong Wongchang
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
- Division of Pharmacology, Department of Pharmaceutical Care, School of Pharmaceutical Sciences, University of Phayao, Phayao, Thailand
| | - Nao Katsurada
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Tatsuaki Tsuruyama
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
- Department of Medical Technology and Sciences, Graduate School of Health Sciences, Kyoto Tachibana University, Kyoto, 607-8175, Japan
| | - Masaki Tajima
- Division of Integrated High-Order Regulatory Systems, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Yutaka Enomoto
- Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Toshio Kitamura
- Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
- Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, 650-0047, Japan
| | - Masahiro Kawashima
- Department of Breast Surgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Masakazu Toi
- Department of Breast Surgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Koji Yamanoi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Shigeo Hisamori
- Department of Gastrointestinal Surgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Kazutaka Obama
- Department of Gastrointestinal Surgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Varodom Charoensawan
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Integrative Computational BioScience (ICBS) Center, Mahidol University, Nakhon Pathom, 73170, Thailand
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand
| | - Dean Thumkeo
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan.
- AMED-FORCE, Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan.
- Foundation for Biomedical Research and Innovation at Kobe, Kobe, 650-0047, Japan.
| |
Collapse
|
3
|
Lin Y, Sun N, Liu D, Yang X, Dong Y, Jiang C. COX-2/PTGS2-targeted herbal-derived oligonucleotide drug HQi-sRNA-2 was effective in spontaneous mouse lung cancer model. IUBMB Life 2024; 76:937-950. [PMID: 39051847 DOI: 10.1002/iub.2858] [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: 11/04/2023] [Accepted: 03/13/2024] [Indexed: 07/27/2024]
Abstract
In 2020, the number of deaths caused by lung cancer worldwide reached 1,796,144, making it the leading cause of cancer-related deaths. Cyclooxygenase-2/prostaglandin endoperoxide synthase 2 (COX-2/PTGS2) is overexpressed in lung cancer, which promotes tumor proliferation, invasion, angiogenesis, and resistance to apoptosis. Here, we report that the oligonucleotide drug HQi-sRNA-2 from Traditional Chinese Medicine Huangqin targeting COX-2/PTGS2 significantly inhibited proliferation, migration, and invasion and induced apoptosis in the human lung cancer cell line NCI-H460. Oral delivery of HQi-sRNA-2 bencaosomes prolonged survival, reduced tumor burden, and maintained weight in a spontaneous mouse lung cancer model. Compared with paclitaxel, HQi-sRNA-2 may be less toxic and have approximately equal efficacy in reducing tumor burden. Our previous studies reported that herbal small RNAs (sRNAs) are functional medical components. Our data suggest that sphingosine (d18:1)-HQi-sRNA-2 bencaosomes, targeting COX-2/PTGS2 and downregulating the PI3K and AKT signaling pathways, may provide novel therapeutics for lung cancer.
Collapse
Affiliation(s)
- Yexuan Lin
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Sun
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dengyuan Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinmeng Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yixin Dong
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chengyu Jiang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Kaufmann M, Vaysse PM, Savage A, Kooreman LFS, Janssen N, Varma S, Ren KYM, Merchant S, Engel CJ, Olde Damink SWM, Smidt ML, Shousha S, Chauhan H, Karali E, Kazanc E, Poulogiannis G, Fichtinger G, Tauber B, Leff DR, Pringle SD, Rudan JF, Heeren RMA, Porta Siegel T, Takáts Z, Balog J. Testing of rapid evaporative mass spectrometry for histological tissue classification and molecular diagnostics in a multi-site study. Br J Cancer 2024; 131:1298-1308. [PMID: 39294437 PMCID: PMC11473823 DOI: 10.1038/s41416-024-02739-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: 10/13/2023] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND While REIMS technology has successfully been demonstrated for the histological identification of ex-vivo breast tumor tissues, questions regarding the robustness of the approach and the possibility of tumor molecular diagnostics still remain unanswered. In the current study, we set out to determine whether it is possible to acquire cross-comparable REIMS datasets at multiple sites for the identification of breast tumors and subtypes. METHODS A consortium of four sites with three of them having access to fresh surgical tissue samples performed tissue analysis using identical REIMS setups and protocols. Overall, 21 breast cancer specimens containing pathology-validated tumor and adipose tissues were analyzed and results were compared using uni- and multivariate statistics on normal, WT and PIK3CA mutant ductal carcinomas. RESULTS Statistical analysis of data from standards showed significant differences between sites and individual users. However, the multivariate classification models created from breast cancer data elicited 97.1% and 98.6% correct classification for leave-one-site-out and leave-one-patient-out cross validation. Molecular subtypes represented by PIK3CA mutation gave consistent results across sites. CONCLUSIONS The results clearly demonstrate the feasibility of creating and using global classification models for a REIMS-based margin assessment tool, supporting the clinical translatability of the approach.
Collapse
Affiliation(s)
- Martin Kaufmann
- Department of Surgery, Queen's University, Kingston, ON, Canada
- Gastrointestinal Diseases Research Unit, Kingston Health Sciences Centre, Kingston, ON, Canada
| | - Pierre-Maxence Vaysse
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, NL, Netherlands
- Department of Surgery, Maastricht University Medical Center + (MUMC+), Maastricht, NL, Netherlands
- Department of Otorhinolaryngology, Head & Neck Surgery, MUMC+, Maastricht, NL, Netherlands
| | - Adele Savage
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Loes F S Kooreman
- Department of Pathology, MUMC+, Maastricht, NL, Netherlands
- GROW School for Oncology and Reproduction, MUMC+, Maastricht, NL, Netherlands
| | - Natasja Janssen
- School of Computing, Queen's University, Kingston, ON, Canada
| | - Sonal Varma
- Department of Pathology, Queen's University, Kingston, ON, Canada
| | - Kevin Yi Mi Ren
- Department of Pathology, Queen's University, Kingston, ON, Canada
| | - Shaila Merchant
- Department of Surgery, Queen's University, Kingston, ON, Canada
| | - Cecil Jay Engel
- Department of Surgery, Queen's University, Kingston, ON, Canada
| | - Steven W M Olde Damink
- Department of Surgery, Maastricht University Medical Center + (MUMC+), Maastricht, NL, Netherlands
- Department of General, Visceral and Transplantation Surgery, RWTH University Hospital Aachen, Aachen, Germany
- NUTRIM School of Nutrition and Translational Research in Metabolism Faculty of Health, Maastricht University, Maastricht, NL, Netherlands
| | - Marjolein L Smidt
- Department of Surgery, Maastricht University Medical Center + (MUMC+), Maastricht, NL, Netherlands
- GROW School for Oncology and Reproduction, MUMC+, Maastricht, NL, Netherlands
| | | | - Hemali Chauhan
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Evdoxia Karali
- Signalling and Cancer Metabolism Team, The Institute of Cancer Research, London, UK
| | - Emine Kazanc
- Signalling and Cancer Metabolism Team, The Institute of Cancer Research, London, UK
| | - George Poulogiannis
- Signalling and Cancer Metabolism Team, The Institute of Cancer Research, London, UK
| | | | - Boglárka Tauber
- Qamcom Research & Technology Central Europe, Budapest, Hungary
| | - Daniel R Leff
- Department of Surgery and Cancer, Biosurgery and Surgical Technology, Imperial College London, London, UK
| | | | - John F Rudan
- Department of Surgery, Queen's University, Kingston, ON, Canada
| | - Ron M A Heeren
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, NL, Netherlands
| | - Tiffany Porta Siegel
- Maastricht MultiModal Molecular Imaging (M4I) Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, NL, Netherlands
| | - Zoltán Takáts
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Júlia Balog
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
- Waters Research Center, Budapest, Hungary.
| |
Collapse
|
5
|
Képes Z, Szabó JP, Kálmán-Szabó I, Sass T, Esze R, Opposits G, Jószai I, Szikra D, Fenyvesi F, Hajdu I, Trencsényi G. 52Mn-labelled Beta-cyclodextrin for Melanoma Imaging: A Proof-of-concept Preclinical Study. In Vivo 2024; 38:2591-2600. [PMID: 39477386 DOI: 10.21873/invivo.13735] [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: 07/04/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 11/07/2024]
Abstract
BACKGROUND/AIM As prostaglandin E2 (PGE2) and its receptors (EP2) are over-expressed on tumor cells and microenvironment, radiolabeled cyclodextrins targeting such biomolecules are valuable vector candidates in molecular cancer diagnostics. Using experimental melanoma models, we evaluated the in vivo imaging behavior of novel Manganese-52-labeled (52Mn) randomly methylated beta-cyclodextrin ([52Mn]Mn-DOTAGA-RAMEB) and compared it with the following well-established tumor-specific probes: melanocortin-1 receptor (MC1-R)-affine [68Ga]Ga-DOTA-NAPamide and PGE2 selective [68Ga]Ga-DOTAGA-RAMEB cyclodextrin. MATERIALS AND METHODS Post-injection of [68Ga]Ga-DOTA-NAPamide, [68Ga]Ga-DOTAGA-RAMEB, and [52Mn]Mn-DOTAGA-RAMEB into MC1-R positive B16F10 melanoma-bearing mice, tumor radio-pharmaceutical uptake was quantified in vivo and ex vivo using preclinical positron emission tomography (PET) and high-performance gamma counter. RESULTS Although all tracers performed well in tumor identification, the highest standardized uptake values were detected in the [68Ga]Ga-DOTA-NAPamide scans. Corresponding to the ex vivo data, meaningful [52Mn]Mn-DOTAGA-RAMEB accumulation 1 h post-injection confirmed the tumor-targeting potential of the tracer. Temporal changes in PGE2/EP2 expression of the neoplasms may explain the significant differences observed between the tumor uptake of the two cyclodextrin probes and that of the 52Mn-labelled compound measured 1 h, 4 h, and 3 days post-injection (p≤0.01, p≤0.05). CONCLUSION Although further pharmacokinetical optimization may be required, 52Mn-labelled cyclodextrin holds potential in melanoma diagnostics and the PET-based longitudinal assessment of tumor-associated PGE2/EP2 expression.
Collapse
Affiliation(s)
- Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary;
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit P Szabó
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ibolya Kálmán-Szabó
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Sass
- Department of Surgery, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Regina Esze
- Division of Metabolic Diseases, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Opposits
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Jószai
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dezső Szikra
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ferenc Fenyvesi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - István Hajdu
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
6
|
Perepechaeva ML, Stefanova NA, Grishanova AY, Kolosova NG. The Expression of Genes CYP1A1, CYP1B1, and CYP2J3 in Distinct Regions of the Heart and Its Possible Contribution to the Development of Hypertension. Biomedicines 2024; 12:2374. [PMID: 39457686 PMCID: PMC11505345 DOI: 10.3390/biomedicines12102374] [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: 09/23/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND It is believed that alterations in the functioning of the cytochrome P450 (CYP), which participates in metabolic transformations of endogenous polyunsaturated fatty acids (PUFAs) (with the formation of cardioprotective or cardiotoxic products), affects the development of age-related cardiovascular diseases and reduces the effectiveness of some cardioselective drugs. For example, CYP2J2 activation or CYP1B1 inhibition protects against the cardiovascular toxicity of anticancer drugs. It is currently unclear whether CYPs capable of metabolizing arachidonic acid and ω-3 PUFAs to vasodilatory and vasoconstrictive derivatives are expressed in all heart regions. METHODS The work was performed on senescence-accelerated OXYS rats featuring elevated blood pressure, OXYSb rats (an OXYS substrain with normal blood pressure), and Wistar rats as a "healthy" control. The mRNA level was determined in the right and left ventricles, the right and left atria, and the aorta of 1-, 3-, and 12-month-old rats. RESULTS We showed that all heart regions express CYPs capable of metabolizing arachidonic acid and ω-3 PUFAs and revealed significant differences between heart regions both in the mRNA level of genes CYP1B1, CYP2J3, and CYP1A1 and in the time course of expression changes with age. CONCLUSIONS We noticed that expression levels of these CYPs in the heart regions and aorta differ between hypertensive OXYS rats, normotensive OXYSb rats, and healthy Wistar rats but could not detect any clear-cut patterns associated with the hypertensive status of OXYS rats.
Collapse
Affiliation(s)
- Maria L. Perepechaeva
- Institute of Molecular Biology and Biophysics, Federal Research Center for Fundamental and Translational Medicine, Timakova Str. 2, Novosibirsk 630060, Russia;
| | - Natalia A. Stefanova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Akad. Lavrentiev Ave., Novosibirsk 630090, Russia; (N.A.S.); (N.G.K.)
| | - Alevtina Y. Grishanova
- Institute of Molecular Biology and Biophysics, Federal Research Center for Fundamental and Translational Medicine, Timakova Str. 2, Novosibirsk 630060, Russia;
| | - Nataliya G. Kolosova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Akad. Lavrentiev Ave., Novosibirsk 630090, Russia; (N.A.S.); (N.G.K.)
| |
Collapse
|
7
|
Mastrogeorgiou M, Chatzikalil E, Theocharis S, Papoudou-Bai A, Péoc'h M, Mobarki M, Karpathiou G. The immune microenvironment of cancer of the uterine cervix. Histol Histopathol 2024; 39:1245-1271. [PMID: 38483012 DOI: 10.14670/hh-18-727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
While several treatment choices exist for cervical cancer, such as surgical therapy, chemotherapy, and radiotherapy, some patients will still show poor prognosis. HPV infection is a principal factor for cervical cancer development, from early inflammation to proliferation, angiogenesis, and neoplastic growth. While HPV T-cell responses exist, the tumor seems to evade the immune system upon its tolerance. The latter suggests the existence of a confluent tumor microenvironment responsible for the evasion tactics employed by the neoplasm. Therefore, novel biomarkers governing prognosis and treatment planning must be developed, with several studies tackling the significance of the tumor microenvironment in the genesis, development, proliferation, and overall response of cervical cancer during neoplastic processes. This review aims to analyze and contemplate the characteristics of the tumor microenvironment and its role in prognosis, progression, evasion, and invasion, including therapeutic outcome and overall survival.
Collapse
Affiliation(s)
- Michail Mastrogeorgiou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Elena Chatzikalil
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Michel Péoc'h
- Department of Pathology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Mousa Mobarki
- Department of Pathology, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
| | - Georgia Karpathiou
- Department of Pathology, University Hospital of Saint-Etienne, Saint-Etienne, France.
| |
Collapse
|
8
|
Yeon Kim S, Tang M, Lu T, Chih SY, Li W. Ferroptosis in glioma therapy: advancements in sensitizing strategies and the complex tumor-promoting roles. Brain Res 2024; 1840:149045. [PMID: 38821335 PMCID: PMC11323215 DOI: 10.1016/j.brainres.2024.149045] [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: 02/27/2024] [Revised: 05/03/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Ferroptosis, an iron-dependent form of non-apoptotic regulated cell death, is induced by the accumulation of lipid peroxides on cellular membranes. Over the past decade, ferroptosis has emerged as a crucial process implicated in various physiological and pathological systems. Positioned as an alternative modality of cell death, ferroptosis holds promise for eliminating cancer cells that have developed resistance to apoptosis induced by conventional therapeutics. This has led to a growing interest in leveraging ferroptosis for cancer therapy across diverse malignancies. Gliomas are tumors arising from glial or precursor cells, with glioblastoma (GBM) being the most common malignant primary brain tumor that is associated with a dismal prognosis. This review provides a summary of recent advancements in the exploration of ferroptosis-sensitizing methods, with a specific focus on their potential application in enhancing the treatment of gliomas. In addition to summarizing the therapeutic potential, this review also discusses the intricate interplay of ferroptosis and its potential tumor-promoting roles within gliomas. Recognizing these dual roles is essential, as they could potentially complicate the therapeutic benefits of ferroptosis. Exploring strategies aimed at circumventing these tumor-promoting roles could enhance the overall therapeutic efficacy of ferroptosis in the context of glioma treatment.
Collapse
Affiliation(s)
- Soo Yeon Kim
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Miaolu Tang
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Tong Lu
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Stephen Y Chih
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA; Medical Scientist Training Program, Penn State College of Medicine, Hershey, PA, USA
| | - Wei Li
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA; Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA; Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
9
|
Liu G, Mu KL, Ran F, Liu JM, Zhou LL, Peng LQ, Feng G, Liu YC, Wei FD, Zhu LL, Zhang XY, Zhang YP, Sun QW. The hemostatic activity and Mechanistic roles of glucosyloxybenzyl 2-isobutylmalate extract (BSCE) from Bletilla striata (Thunb.) Rchb.f. in Inhibiting pulmonary hemorrhage. Heliyon 2024; 10:e38203. [PMID: 39381249 PMCID: PMC11459001 DOI: 10.1016/j.heliyon.2024.e38203] [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: 05/03/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
Background Hemorrhagic events cause numerous deaths annually worldwide, highlighting the urgent need for effective hemostatic drugs. The glucosyloxybenzyl 2-isobutylmalates Control Extract (BSCE) from the orchid plant Bletilla striata (Thunb.) Rchb.f. has demonstrated significant hemostatic activity in both in vitro and in vivo studies. However, the effect and mechanism of BSCE on non-traumatic bleeding remain unclear. Methods Pulmonary hemorrhage was induced in 40 Sprague-Dawley rats by administering Zingiber officinale Roscoe. for 14 days. These rats were then randomly divided into five groups: model (Mod), positive control (YNBY), and BSCE low, medium, and high-dose groups. An additional 8 rats served as the control group (Con). The BSCE groups received different doses of BSCE for 10 days, while the YNBY group received Yunnan Baiyao suspension. The effects on body weight, food and water intake, red blood cell count (RBC), hemoglobin concentration (HGB), lung tissue pathology, platelet count, coagulation parameters, and fibrinolytic system markers were evaluated. Network pharmacology and molecular docking analyses were also conducted to identify potential targets and pathways involved in BSCE's effects. Results BSCE treatment significantly improved body weight, food intake, and water consumption in rats with pulmonary hemorrhage. RBC and HGB levels increased significantly in the BSCE medium and high-dose groups compared to the Mod group (P < 0.05). Pathological examination revealed that BSCE reduced lung tissue hemorrhage and inflammation, with improvements in alveolar structure. BSCE also positively affected platelet count, thrombin time (TT), activated partial thromboplastin time (APTT), fibrinogen (FIB) levels, and fibrinolytic markers (D-dimer, PAI-1, and t-PA). Network pharmacology and molecular docking identified key targets such as MMPs, CASPs, and pathways including IL-17 and TNF signaling, suggesting BSCE's involvement in hemostasis and anti-inflammatory processes. Conclusions BSCE exhibits significant hemostatic and protective effects on Z.officinale-induced pulmonary hemorrhage in rats by improving hematological parameters, reducing lung tissue damage, and modulating the coagulation and fibrinolytic systems. The study provides evidence supporting the potential of BSCE as a therapeutic agent for hemorrhagic diseases, with its efficacy linked to multi-target and multi-pathway interactions.
Collapse
Affiliation(s)
| | | | - Fei Ran
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Jin-mei Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Ling-li Zhou
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Le-qiang Peng
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Guo Feng
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Yu-chen Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Fu-dao Wei
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Ling-li Zhu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Xin-yue Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Yong-ping Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Qing-wen Sun
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| |
Collapse
|
10
|
Xu X, Zhang Y, Meng C, Zheng W, Wang L, Zhao C, Luo F. Nanozymes in cancer immunotherapy: metabolic disruption and therapeutic synergy. J Mater Chem B 2024; 12:9111-9143. [PMID: 39177061 DOI: 10.1039/d4tb00769g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Over the past decade, there has been a growing emphasis on investigating the role of immunotherapy in cancer treatment. However, it faces challenges such as limited efficacy, a diminished response rate, and serious adverse effects. Nanozymes, a subset of nanomaterials, demonstrate boundless potential in cancer catalytic therapy for their tunable activity, enhanced stability, and cost-effectiveness. By selectively targeting the metabolic vulnerabilities of tumors, they can effectively intensify the destruction of tumor cells and promote the release of antigenic substances, thereby eliciting immune clearance responses and impeding tumor progression. Combined with other therapies, they synergistically enhance the efficacy of immunotherapy. Hence, a large number of metabolism-regulating nanozymes with synergistic immunotherapeutic effects have been developed. This review summarizes recent advancements in cancer immunotherapy facilitated by nanozymes, focusing on engineering nanozymes to potentiate antitumor immune responses by disturbing tumor metabolism and performing synergistic treatment. The challenges and prospects in this field are outlined. We aim to provide guidance for nanozyme-mediated immunotherapy and pave the way for achieving durable tumor eradication.
Collapse
Affiliation(s)
- Xiangrui Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yaowen Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chijun Meng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wenzhuo Zheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lingfeng Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chenyi Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Feng Luo
- Department of Prosthodontics, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu 610041, China.
| |
Collapse
|
11
|
Musa MS, Miah MS, Munni YA, Patwary MAM, Kazi M, Matin MM. Synthesis and elucidation of strained galactopyronose esters as selective cyclooxygenase-2 inhibitor: a comprehensive computational approach. RSC Adv 2024; 14:30469-30481. [PMID: 39318455 PMCID: PMC11421415 DOI: 10.1039/d4ra03520h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024] Open
Abstract
Cyclooxygenase-2 (COX-2) is critically implicated in various pathologies, including inflammation, cancer, disorders involving the nervous system, and multidrug resistance. In both academic and pharmaceutical research, the development of COX-2 selective drugs as anti-inflammatory and anti-tumor therapeutics is a key focus. Traditional nonsteroidal anti-inflammatory drugs (NSAIDs) have ulcerogenic, gastrointestinal adverse effects, and myocardial infarction risk, which resulted in their limited applications. In response to this challenge, we synthesized a series of glycoconjugates featuring six-membered sugar rings and acyl chains of varying lengths attached at the C-6 position. Using molecular docking techniques, we identified galactose esters with optimal acyl chain lengths that selectively and effectively bind to the active site of COX-2 over COX-1. These compounds exhibited enhanced binding affinity and superior inhibition constants (pK i) for COX-2, thereby offering selective inhibition with potentially reduced ulcerogenic risks, as COX-1 inhibition is thought to contribute to these side effects. The molecular docking study identified two potential compounds, G6 and G8, which were validated via MD simulation for the assessment of their stability and were compared to the complex of the standard drugs, aspirin and rofecoxib. In addition, compound structures were optimized using the DFT method under the B3LYP/6-31+g(d,p) basis set to study their physio-spectral properties, frontier molecular orbitals (HOMO-LUMO), and their energy gap that correlates to their reactivity and stability. ADMET, drug-likeness, and PASS analyses were also carried out to assess their drug-ability and toxicity profiling.
Collapse
Affiliation(s)
- Mohammed Sakib Musa
- Department of Applied Chemistry and Chemical Engineering, Faculty of Science, University of Chittagong Chittagong 4331 Bangladesh
| | - Md Sopon Miah
- Bioorganic and Medicinal Chemistry Laboratory, Department of Chemistry, Faculty of Science, University of Chittagong Chittagong 4331 Bangladesh +880 1716 839689
| | - Yeasmin Akter Munni
- Department of Anatomy, College of Medicine, Dongguk University Gyeongju 38066 Republic of Korea
| | | | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University P. O. Box 2457 Riyadh 11451 Saudi Arabia
| | - Mohammed Mahbubul Matin
- Bioorganic and Medicinal Chemistry Laboratory, Department of Chemistry, Faculty of Science, University of Chittagong Chittagong 4331 Bangladesh +880 1716 839689
| |
Collapse
|
12
|
Yadav V, Singh T, Sharma D, Garg VK, Chakraborty P, Ghatak S, Satapathy SR. Unraveling the Regulatory Role of HuR/microRNA Axis in Colorectal Cancer Tumorigenesis. Cancers (Basel) 2024; 16:3183. [PMID: 39335155 PMCID: PMC11430344 DOI: 10.3390/cancers16183183] [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: 08/07/2024] [Revised: 09/04/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Colorectal cancer (CRC) remains a significant global health burden with high incidence and mortality. MicroRNAs (miRNAs) are small non-protein coding transcripts, conserved throughout evolution, with an important role in CRC tumorigenesis, and are either upregulated or downregulated in various cancers. RNA-binding proteins (RBPs) are known as essential regulators of miRNA activity. Human antigen R (HuR) is a prominent RBP known to drive tumorigenesis with a pivotal role in CRC. In this review, we discuss the regulatory role of the HuR/miRNA axis in CRC. Interestingly, miRNAs can directly target HuR, altering its expression and activity. However, HuR can also stabilize or degrade miRNAs, forming complex feedback loops that either activate or block CRC-associated signaling pathways. Dysregulation of the HuR/miRNA axis contributes to CRC initiation and progression. Additionally, HuR-miRNA regulation by other small non-coding RNAs, circular RNA (circRNAs), or long-non-coding RNAs (lncRNAs) is also explored here. Understanding this HuR-miRNA interplay could reveal novel biomarkers with better diagnostic or prognostic accuracy.
Collapse
Affiliation(s)
- Vikas Yadav
- Department of Translational Medicine, Clinical Research Centre, Lund University, 221 00 Malmö, Sweden;
| | - Tejveer Singh
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, University of Delhi, New Delhi 110021, India; (T.S.); (D.S.)
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences (INMAS-DRDO), New Delhi 110054, India
| | - Deepika Sharma
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, University of Delhi, New Delhi 110021, India; (T.S.); (D.S.)
| | - Vivek Kumar Garg
- Department of Medical Lab Technology, Chandigarh University, Gharuan, Mohali 140413, Punjab, India;
| | - Payel Chakraborty
- Amity Institute of Biotechnology, Amity University Kolkata, Kolkata 700135, West Bengal, India; (P.C.); (S.G.)
| | - Souvik Ghatak
- Amity Institute of Biotechnology, Amity University Kolkata, Kolkata 700135, West Bengal, India; (P.C.); (S.G.)
| | - Shakti Ranjan Satapathy
- Department of Translational Medicine, Clinical Research Centre, Lund University, 221 00 Malmö, Sweden;
| |
Collapse
|
13
|
Calabrese C, Miserocchi G, De Vita A, Spadazzi C, Cocchi C, Vanni S, Gabellone S, Martinelli G, Ranallo N, Bongiovanni A, Liverani C. Lipids and adipocytes involvement in tumor progression with a focus on obesity and diet. Obes Rev 2024:e13833. [PMID: 39289899 DOI: 10.1111/obr.13833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
The adipose tissue is a complex organ that can play endocrine, metabolic, and immune regulatory roles in cancer. In particular, adipocytes provide metabolic substrates for cancer cell proliferation and produce signaling molecules that can stimulate cell adhesion, migration, invasion, angiogenesis, and inflammation. Cancer cells, in turn, can reprogram adipocytes towards a more inflammatory state, resulting in a vicious cycle that fuels tumor growth and evolution. These mechanisms are enhanced in obesity, which is associated with the risk of developing certain tumors. Diet, an exogenous source of lipids with pro- or anti-inflammatory functions, has also been connected to cancer risk. This review analyzes how adipocytes and lipids are involved in tumor development and progression, focusing on the relationship between obesity and cancer. In addition, we discuss how diets with varying lipid intakes can affect the disease outcomes. Finally, we introduce novel metabolism-targeted treatments and adipocyte-based therapies in oncology.
Collapse
Affiliation(s)
- Chiara Calabrese
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giacomo Miserocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandro De Vita
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Spadazzi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Claudia Cocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Silvia Vanni
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sofia Gabellone
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Martinelli
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Nicoletta Ranallo
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alberto Bongiovanni
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Liverani
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
14
|
Zhu X, Huang H, Zou M, Luo H, Liu T, Zhu S, Ye B. Identification of circulating metabolites linked to the risk of breast cancer: a mendelian randomization study. Front Pharmacol 2024; 15:1442723. [PMID: 39323635 PMCID: PMC11422656 DOI: 10.3389/fphar.2024.1442723] [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: 06/02/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Objective This study aimed to investigate potential causal relationships between circulating metabolites and breast cancer risk using Mendelian randomization (MR) analysis. Materials and Methods Summary-level genome-wide association study (GWAS) datasets for 249 circulating metabolites were obtained from the UK Biobank. GWAS datasets for estrogen receptor-positive (ER+) and estrogen receptor-negative (ER-) breast cancer were acquired from previous studies based on the Combined Oncoarray. Instrumental variables (IVs) were selected from single nucleotide polymorphisms (SNPs) associated with circulating metabolites, and MR analyses were conducted using the inverse-variance weighted (IVW) method as the primary analysis, with additional sensitivity analyses using other MR methods. Odds ratios (OR) and 95% confidence interval (CI) were used to estimate the association of circulating metabolites with breast cancer risk. Results The IVW analysis revealed significant causal relationships between 79 circulating metabolites and ER + breast cancer risk, and 10 metabolites were significantly associated with ER-breast cancer risk. Notably, acetate (OR = 1.12, P = 0.03), HDL cholesterol (OR = 1.09, P < 0.001), ration of omega-6 fatty acids to total fatty acids ratio (OR = 1.09, P = 0.01), and phospholipids in large LDL (OR = 1.09, P < 0.001) were linked to an increased risk of ER + breast cancer, while linoleic acid (OR = 0.91, P < 0.001) monounsaturated fatty acids (OR = 0.91, P < 0.001), and total lipids in LDL (OR = 0.91, P < 0.001) were associated with a decreased risk. In ER-breast cancer, glycine, citrate, HDL cholesterol, cholesteryl esters in HDL, cholesterol to total lipids ratio in very large HDL, and cholesterol in large LDL were associated with an increased risk, while the free cholesterol to total lipids in very large HDL was linked to a decreased risk. Conclusion This MR approach underscores aberrant lipid metabolism as a key process in breast tumorigenesis, and may inform future prevention and treatment strategies. To further elucidate the underlying mechanisms and explore the potential clinical implications, additional research is warranted to validate the observed associations in this study.
Collapse
Affiliation(s)
- Xiaosheng Zhu
- Department of Radiation, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Huai Huang
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Mengjie Zou
- Department of Nephrology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Honglin Luo
- Institute of Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Tianqi Liu
- Department of General Surgery, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Shaoliang Zhu
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Bin Ye
- Department of Radiation, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
15
|
Li H, Fang G, Tian W, Liao Y, Xiang J, Hu Y, Luo L. Asiatic acid induces lung cancer toxicity by triggering SRC-mediated ferroptosis. Toxicol Appl Pharmacol 2024; 492:117097. [PMID: 39251043 DOI: 10.1016/j.taap.2024.117097] [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: 05/31/2024] [Revised: 08/12/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Ferroptosis is a recently discovered form of regulated cell death that shows promise as a novel approach for inducing tumor cell death in cancer treatment, with significant research potential. Asiatic acid (AA), a key component of the traditional Chinese medicine Centella asiatica, has been identified as having potential therapeutic benefits for various diseases, particularly cancer. Non-small cell lung cancer (NSCLC) is a challenging and prevalent form of cancer to treat. In our study, we utilized network pharmacology, molecular docking, and experimental methods to investigate the potential of AA in treating NSCLC and to elucidate its role in inhibiting cancer through the ferroptosis pathway. Through network pharmacology analysis, we identified that AA targets the core NSCLC protein SRC through the ferroptosis pathway. Our experiments demonstrated that treatment with AA led to increased iron accumulation, mitochondrial membrane potential, and expression of ferroptosis markers glutathione peroxidase 4 (GPX4), ferritin heavy chain 1 (FTH1), and acyl-CoA synthetase long chain family member 4 (ACSL4) in NSCLC cells, confirming the induction of ferroptosis. In conclusion, AA has the potential to target SRC and induce NSCLC cell death through the ferroptosis pathway, offering a promising approach for cancer treatment.
Collapse
Affiliation(s)
- Huizhen Li
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Guixuan Fang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Wen Tian
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Yinglin Liao
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Jing Xiang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Yingying Hu
- Department of Pathophysiology, Guangdong Medical University, Zhanjiang 524002, Guangdong, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
16
|
Guo Y, Wang S, Wu X, Zhao R, Chang S, Ma C, Song S, Zeng S. Multi-Omics Reveals the Role of Arachidonic Acid Metabolism in the Gut-Follicle Axis for the Antral Follicular Development of Holstein Cows. Int J Mol Sci 2024; 25:9521. [PMID: 39273467 PMCID: PMC11395146 DOI: 10.3390/ijms25179521] [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: 07/06/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
In vitro embryonic technology is crucial for improving farm animal reproduction but is hampered by the poor quality of oocytes and insufficient development potential. This study investigated the relationships among changes in the gut microbiota and metabolism, serum features, and the follicular fluid metabolome atlas. Correlation network maps were constructed to reveal how the metabolites affect follicular development by regulating gene expression in granulosa cells. The superovulation synchronization results showed that the number of follicle diameters from 4 to 8 mm, qualified oocyte number, cleavage, and blastocyst rates were improved in the dairy heifers (DH) compared with the non-lactating multiparous dairy cows (NDC) groups. The gut microbiota was decreased in Rikenellaceae_RC9_gut_group, Alistipes, and Bifidobacterium, but increased in Firmicutes, Cyanobacteria, Fibrobacterota, Desulfobacterota, and Verrucomicrobiota in the NDC group, which was highly associated with phospholipid-related metabolites of gut microbiota and serum. Metabolomic profiling of the gut microbiota, serum, and follicular fluid further demonstrated that the co-metabolites were phosphocholine and linoleic acid. Moreover, the expression of genes related to arachidonic acid metabolism in granulosa cells was significantly correlated with phosphocholine and linoleic acid. The results in granulosa cells showed that the levels of PLCB1 and COX2, participating in arachidonic acid metabolism, were increased in the DH group, which improved the concentrations of PGD2 and PGF2α in the follicular fluid. Finally, the expression levels of apoptosis-related proteins, cytokines, and steroidogenesis-related genes in granulosa cells and the concentrations of steroid hormones in follicular fluid were determinants of follicular development. According to our results, gut microbiota-related phosphocholine and linoleic acid participate in arachidonic acid metabolism in granulosa cells through the gut-follicle axis, which regulates follicular development. These findings hold promise for enhancing follicular development and optimizing oocyte quality in subfertile dairy cows.
Collapse
Affiliation(s)
- Yajun Guo
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shiwei Wang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xuan Wu
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Rong Zhao
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Siyu Chang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Chen Ma
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shuang Song
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shenming Zeng
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
17
|
Liang XY, Wang Y, Zhu YW, Zhang YX, Yuan H, Liu YF, Jin YQ, Gao W, Ren ZG, Ji XY, Wu DD. Role of hydrogen sulfide in dermatological diseases. Nitric Oxide 2024; 150:18-26. [PMID: 38971520 DOI: 10.1016/j.niox.2024.07.001] [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: 03/28/2024] [Revised: 06/30/2024] [Accepted: 07/04/2024] [Indexed: 07/08/2024]
Abstract
Hydrogen sulfide (H2S), together with carbon monoxide (CO) and nitric oxide (NO), is recognized as a vital gasotransmitter. H2S is biosynthesized by enzymatic pathways in the skin and exerts significant physiological effects on a variety of biological processes, such as apoptosis, modulation of inflammation, cellular proliferation, and regulation of vasodilation. As a major health problem, dermatological diseases affect a large proportion of the population every day. It is urgent to design and develop effective drugs to deal with dermatological diseases. Dermatological diseases can arise from a multitude of etiologies, including neoplastic growth, infectious agents, and inflammatory processes. The abnormal metabolism of H2S is associated with many dermatological diseases, such as melanoma, fibrotic diseases, and psoriasis, suggesting its therapeutic potential in the treatment of these diseases. In addition, therapies based on H2S donors are being developed to treat some of these conditions. In the review, we discuss recent advances in the function of H2S in normal skin, the role of altering H2S metabolism in dermatological diseases, and the therapeutic potential of diverse H2S donors for the treatment of dermatological diseases.
Collapse
Affiliation(s)
- Xiao-Yi Liang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Yan Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Yi-Wen Zhu
- School of Clinical Medicine, Henan University, Kaifeng, Henan, 475004, China
| | - Yan-Xia Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Hang Yuan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Ya-Fang Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Yu-Qing Jin
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Wei Gao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
| | - Zhi-Guang Ren
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China; Kaifeng Key Laboratory of Infectious Diseases and Biosafety, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China; Kaifeng Key Laboratory of Infectious Diseases and Biosafety, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China; Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan 450064, China.
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China; Kaifeng Key Laboratory of Infectious Diseases and Biosafety, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China; Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
18
|
Buglakova E, Ekelöf M, Schwaiger-Haber M, Schlicker L, Molenaar MR, Shahraz M, Stuart L, Eisenbarth A, Hilsenstein V, Patti GJ, Schulze A, Snaebjornsson MT, Alexandrov T. Spatial single-cell isotope tracing reveals heterogeneity of de novo fatty acid synthesis in cancer. Nat Metab 2024; 6:1695-1711. [PMID: 39251875 PMCID: PMC11422168 DOI: 10.1038/s42255-024-01118-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 07/26/2024] [Indexed: 09/11/2024]
Abstract
While heterogeneity is a key feature of cancer, understanding metabolic heterogeneity at the single-cell level remains a challenge. Here we present 13C-SpaceM, a method for spatial single-cell isotope tracing that extends the previously published SpaceM method with detection of 13C6-glucose-derived carbons in esterified fatty acids. We validated 13C-SpaceM on spatially heterogeneous models using liver cancer cells subjected to either normoxia-hypoxia or ATP citrate lyase depletion. This revealed substantial single-cell heterogeneity in labelling of the lipogenic acetyl-CoA pool and in relative fatty acid uptake versus synthesis hidden in bulk analyses. Analysing tumour-bearing brain tissue from mice fed a 13C6-glucose-containing diet, we found higher glucose-dependent synthesis of saturated fatty acids and increased elongation of essential fatty acids in tumours compared with healthy brains. Furthermore, our analysis uncovered spatial heterogeneity in lipogenic acetyl-CoA pool labelling in tumours. Our method enhances spatial probing of metabolic activities in single cells and tissues, providing insights into fatty acid metabolism in homoeostasis and disease.
Collapse
Affiliation(s)
- Elena Buglakova
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Cell Biology and Biophysics Unit, EMBL, Heidelberg, Germany
| | - Måns Ekelöf
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Michaela Schwaiger-Haber
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Lisa Schlicker
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Martijn R Molenaar
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Mohammed Shahraz
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Lachlan Stuart
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Andreas Eisenbarth
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Volker Hilsenstein
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Gary J Patti
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
| | - Marteinn T Snaebjornsson
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
| | - Theodore Alexandrov
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
- Metabolomics Core Facility, EMBL, Heidelberg, Germany.
- Molecular Medicine Partnership Unit, EMBL and Heidelberg University, Heidelberg, Germany.
- BioStudio, BioInnovation Institute, Copenhagen, Denmark.
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
19
|
Patrignani P, Contursi A, Tacconelli S, Steinhilber D. The future of pharmacology and therapeutics of the arachidonic acid cascade in the next decade: Innovative advancements in drug repurposing. Front Pharmacol 2024; 15:1472396. [PMID: 39268466 PMCID: PMC11390530 DOI: 10.3389/fphar.2024.1472396] [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: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Many drugs can act on multiple targets or disease pathways, regardless of their original purpose. Drug repurposing involves reevaluating existing compounds for new medical uses. This can include repositioning approved drugs, redeveloping unapproved drugs, or repurposing any chemical, nutraceutical, or biotherapeutic product for new applications. Traditional drug development is slow, expensive, and has high failure rates. Drug repurposing can speed up the process, costing less and saving time. This approach can save 6-7 years of early-stage research time. Drug repurposing benefits from existing compounds with optimized structures and approved for clinical use with associated structure-activity relationship publications, supporting the development of new effective compounds. Drug repurposes can now utilize advanced in silico screening enabled by artificial intelligence (AI) and sophisticated tissue and organ-level in vitro models. These models more accurately replicate human physiology and improve the selection of existing drugs for further pre-clinical testing and, eventually, clinical trials for new indications. This mini-review discusses some examples of drug repurposing and novel strategies for further development of compounds for targets of the arachidonic acid cascade. In particular, we will delve into the prospect of repurposing antiplatelet agents for cancer prevention and addressing the emerging noncanonical functionalities of 5-lipoxygenase, potentially for leukemia therapy.
Collapse
Affiliation(s)
- Paola Patrignani
- Systems Pharmacology and Translational Therapeutics Laboratory, The Center for Advanced Studies and Technology (CAST), "G. d' Annunzio" University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, "G. d Annunzio" University Medical School, Chieti, Italy
| | - Annalisa Contursi
- Systems Pharmacology and Translational Therapeutics Laboratory, The Center for Advanced Studies and Technology (CAST), "G. d' Annunzio" University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, "G. d Annunzio" University Medical School, Chieti, Italy
| | - Stefania Tacconelli
- Systems Pharmacology and Translational Therapeutics Laboratory, The Center for Advanced Studies and Technology (CAST), "G. d' Annunzio" University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, "G. d Annunzio" University Medical School, Chieti, Italy
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
20
|
Chen T, Liu J, Wang C, Wang Z, Zhou J, Lin J, Mao J, Pan T, Wang J, Xu H, He X, Wu D, Liu Z. ALOX5 contributes to glioma progression by promoting 5-HETE-mediated immunosuppressive M2 polarization and PD-L1 expression of glioma-associated microglia/macrophages. J Immunother Cancer 2024; 12:e009492. [PMID: 39142719 PMCID: PMC11332009 DOI: 10.1136/jitc-2024-009492] [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] [Accepted: 07/30/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Oxylipin metabolism plays an essential role in glioma progression and immune modulation in the tumor microenvironment. Lipid metabolic reprogramming has been linked to macrophage remodeling, while the understanding of oxylipins and their catalyzed enzymes lipoxygenases in the regulation of glioma-associated microglia/macrophages (GAMs) remains largely unexplored. METHODS To explore the pathophysiological relevance of oxylipin in human glioma, we performed Ultra-high performance liquid chromatography-MS/MS (UHPLC-MS/MS) analysis in human glioma and non-tumor brain tissues. To comprehensively investigate the role of arachidonate lipoxygenase 5 (ALOX5) in glioma, we performed in vivo bioluminescent imaging, immunofluorescence staining and flow cytometry analysis on tumors from orthotopic glioma-bearing mice. We developed an ALOX5-targeted nanobody, and tested its anti-glioma efficacy of combination therapy with α-programmed cell death protein-1 (PD-1). RESULTS In this study, we found that ALOX5 and its oxylipin 5-hydroxyeicosatetraenoic acid (5-HETE) are upregulated in glioma, accumulating programmed death-ligand 1 (PD-L1)+ M2-GAMs and orchestrating an immunosuppressive tumor microenvironment. Mechanistically, 5-HETE derived from ALOX5-overexpressing glioma cells, promotes GAMs migration, PD-L1 expression, and M2 polarization by facilitating nuclear translocation of nuclear factor erythroid 2-related factor 2. Additionally, a nanobody targeting ALOX5 is developed that markedly suppresses 5-HETE efflux from glioma cells, attenuates M2 polarization of GAMs, and consequently ameliorates glioma progression. Furthermore, the combination therapy of the ALOX5-targeted nanobody plus α-PD-1 exhibits superior anti-glioma efficacy. CONCLUSIONS Our findings reveal a pivotal role of the ALOX5/5-HETE axis in regulating GAMs and highlight the ALOX5-targeted nanobody as a potential therapeutic agent, which could potentiate immune checkpoint therapy for glioma.
Collapse
Affiliation(s)
- Tao Chen
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Jiangang Liu
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Chenci Wang
- Department of Oncology, Funan County People's Hospital, Fuyang, Anhui, China
| | - Zhengwei Wang
- Department of Neurosurgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Jiayi Zhou
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jiani Lin
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Jie Mao
- Department of Neurosurgery, Longgang Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Tingzheng Pan
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Jianwei Wang
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Hongchao Xu
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Xiaosheng He
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Dinglan Wu
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Zhuohao Liu
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| |
Collapse
|
21
|
Xu H, Chen Y, Tong H, Chen L, Morisseau C, Zhou Z, Zhuang J, Song C, Cai P, Liu Z, Hammock BD, Chen G. Design and Synthesis of sEH/HDAC6 Dual-Targeting Inhibitors for the Treatment of Inflammatory Pain. J Med Chem 2024; 67:12887-12911. [PMID: 39033411 PMCID: PMC11412156 DOI: 10.1021/acs.jmedchem.4c00847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Soluble epoxide hydrolase (sEH) and HDAC6 mediate the NF-κB pathway in inflammatory responses, and their inhibitors exhibit powerful anti-inflammatory and analgesic activities in treating both inflammation and pain. Therefore, a series of dual-targeting inhibitors containing urea or squaramide and hydroxamic acid moieties were designed and synthesized, and their role as a new sEH/HDAC6 dual-targeting inhibitor in inflammatory pain was evaluated in a formalin-induced mice model and a xylene-induced mouse ear swelling model. Among them, compounds 28g and 28j showed the best inhibitory and selectivity of sEH and HDAC6. Compound 28g had satisfactory pharmacokinetic characteristics in rats. Following administration at 30 mg/kg, compound 28g exhibited more effective analgesic activity than either an sEH inhibitor (GL-B437) or an HDAC6 inhibitor (Rocilinostat) alone and coadministration of both inhibitors. Thus, these novel sEH/HDAC6 dual-targeting inhibitors exhibited powerful analgesic activity in nociceptive behavior and are worthy of further development.
Collapse
Affiliation(s)
- Huashen Xu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yuanguang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hua Tong
- Liaoning Key Laboratory of Targeting Drugs for Hematological Malignancies, Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lu Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Christophe Morisseau
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, California 95616, United States
| | - Zijian Zhou
- Liaoning Key Laboratory of Targeting Drugs for Hematological Malignancies, Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Junning Zhuang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chuqiao Song
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Pengcheng Cai
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhongbo Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bruce D Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, California 95616, United States
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
22
|
Luo M, He N, Xu Q, Wen Z, Wang Z, Zhao J, Liu Y. Roles of prostaglandins in immunosuppression. Clin Immunol 2024; 265:110298. [PMID: 38909972 DOI: 10.1016/j.clim.2024.110298] [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: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/21/2024] [Indexed: 06/25/2024]
Abstract
Prostaglandins (PGs) play a crucial and multifaceted role in various physiological processes such as intercellular signaling, inflammation regulation, neurotransmission, vasodilation, vasoconstriction, and reproductive functions. The diversity and biological significance of these effects are contingent upon the specific types or subtypes of PGs, with each PG playing a crucial role in distinct physiological and pathological processes. Particularly within the immune system, PGs are essential in modulating the function of immune cells and the magnitude and orientation of immune responses. Hence, a comprehensive comprehension of the functions PG signaling pathways in immunosuppressive regulation holds substantial clinical relevance for disease prevention and treatment strategies. The manuscript provides a review of recent developments in PG signaling in immunosuppressive regulation. Furthermore, the potential clinical applications of PGs in immunosuppression are also discussed. While research into the immunosuppressive effects of PGs required further exploration, targeted therapies against their immunosuppressive pathways might open new avenues for disease prevention and treatment.
Collapse
Affiliation(s)
- Minjie Luo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Nina He
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Qing Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Zhongchi Wen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Ziqin Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China
| | - Jie Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China.
| | - Ying Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha 410008, Hunan, China; National Medicine Functional Experimental Teaching Center, Changsha 410008, Hunan, China.
| |
Collapse
|
23
|
Zhou Y, Chen H, Yan J, Yao Q, Kong C, Peng Y, Xiao S, Yang J. FOXA2 Activates RND1 to Regulate Arachidonic Acid Metabolism Pathway and Suppress Cisplatin Resistance in Lung Squamous Cell Carcinoma. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e13814. [PMID: 39129202 PMCID: PMC11317498 DOI: 10.1111/crj.13814] [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: 01/21/2024] [Revised: 04/09/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND The primary cause of cancer-related fatalities globally is lung cancer. Although the chemotherapy drug cisplatin (DDP) has brought certain benefits to patients, the rapid development of drug resistance has greatly hindered treatment success. METHODS We used the lung squamous cell carcinoma (LUSC) mRNA data set to explore the differentially expressed gene (RND1) in LUSC and detected RND1 expression in LUSC cells and DDP-resistant cells by qRT-PCR. Meanwhile, we performed abnormal expression treatment on RND1 and conducted CCK8, colony formation, and flow cytometry to evaluate the impact of RND1 expression on cell proliferation, apoptosis, and DDP resistance. In addition, we analyzed metabolism pathways involving RND1 using GSEA. We also used online tools such as hTFtarget and JASPAR to screen for the upstream transcription factor FOXA2 of RND1 and verified their relationship through CHIP and dual luciferase experiments. Finally, we validated the role of FOXA2-RND1 in DDP resistance in LUSC through the above experiments. RESULTS RND1 was downregulated in LUSC, and overexpression of RND1 repressed proliferation and DDP resistance of LUSC cells and facilitated cell apoptosis. RND1 modulated the arachidonic acid (AA) metabolism pathway, and FOXA2 positively manipulated RND1 expression. By activating FOXA2, stabilizing RND1, and regulating AA levels, the sensitivity of LUSC cells to DDP could be enhanced. CONCLUSION Our study suggested that FOXA2 positively modulated the RND1-AA pathway, which repressed the resistance of LUSC cells to DDP.
Collapse
Affiliation(s)
- Yafu Zhou
- Department of Thoracic SurgeonsHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - Huiguo Chen
- Department of Thoracic SurgeonsHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - Jianhua Yan
- Department of Thoracic SurgeonsHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - Qi Yao
- Department of Thoracic SurgeonsHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - Chunchu Kong
- Department of RespiratoryHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - You Peng
- Department of GeriatricHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - Shengying Xiao
- Department of OncologyHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| | - Jinsong Yang
- Department of Thoracic SurgeonsHunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University)ChangshaChina
| |
Collapse
|
24
|
Suska K, Piotrowski M, Fichna J. Lipid biomarkers in colorectal cancer, with particular emphasis on exosomes - current status and future inferences. Expert Rev Gastroenterol Hepatol 2024; 18:441-456. [PMID: 39192805 DOI: 10.1080/17474124.2024.2393180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/16/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024]
Abstract
INTRODUCTION Colorectal cancer (CRC) is one of the most deadly cancers on a global scale. Diagnosis of CRC is challenging and it is often detected at a late stage. Identification of relevant biomarkers could lead to the development of effective diagnostic methods for CRC. AREAS COVERED We reviewed the literature on lipid (including exosomal) biomarkers that have the potential to become common, minimally invasive and effective diagnostic tools for CRC. We showed that differences in lipid levels (single compounds and entire panels) make it possible to classify patients into diseased or healthy groups, determine the stage of CRC, as well as accompanying inflammation and immune reactions associated with tumorigenesis. We also discussed exosomes which are important components of the tumor microenvironment that influence tumor progression and for which only a small number of studies were conducted so far in this area. EXPERT OPINION A rapid development in the field of lipid-based biomarkers, including exosomal lipid biomarkers, is expected as growing evidence shows their potential application and good accuracy. However, one of the major issues that needs to be addressed within this topic is to translate findings into a noninvasive and versatile diagnostic test robustly validated in clinical conditions.
Collapse
Affiliation(s)
- Kinga Suska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Marcin Piotrowski
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | | |
Collapse
|
25
|
Gu W, Zeng D, Zhang C. Discovering the effect of combination of celecoxib and sorafenib on hepatocellular carcinoma. Discov Oncol 2024; 15:321. [PMID: 39083127 PMCID: PMC11291820 DOI: 10.1007/s12672-024-01203-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/29/2024] [Indexed: 08/03/2024] Open
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is a common and fatal cancer, and its molecular mechanisms are still not fully understood. This study aimed to explore the potential molecular mechanisms and immune infiltration characteristics of celecoxib combined with sorafenib in the treatment of HCC by analyzing the differentially expressed genes (DEGs) from the GSE45340 dataset in the GEO database and identifying key genes. METHODS The GSE45340 dataset was downloaded from the GEO database, and DEGs were screened using GEO2R, and visualization and statistical analysis were performed. Metascape was used to perform functional annotation and protein-protein interaction network analysis of DEGs. The immune infiltration was analyzed using the TIMER database, and the expression of key genes and their relationship with patient survival were analyzed and verified using the UALCAN database. RESULTS A total of 2181 DEGs were screened through GEO2R analysis, and heat maps were drawn for the 50 genes with the highest expression. Metascape was used for enrichment analysis, and the enrichment results of KEGG and GO and the PPI network were obtained, and 44 core genes were screened. Analysis of the TIMER database found that 12 genes were closely related to tumor immune infiltration. UALCAN analysis further verified the differential expression of these genes in HCC and was closely related to the overall survival of patients. CONCLUSIONS Through comprehensive bioinformatics analysis, this study identified a group of key genes related to the treatment of HCC with celecoxib combined with sorafenib. These genes play an important role in tumor immune infiltration and patient survival, providing important clues for further studying the molecular mechanism of HCC and developing potential therapeutic targets.
Collapse
Affiliation(s)
- Wang Gu
- Hepatological Surgery Department, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei City, 230032, Anhui Province, China
| | - Dongyun Zeng
- Clinicopathological Diagnosis and Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise, China
| | - Chao Zhang
- Hepatological Surgery Department, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei City, 230032, Anhui Province, China.
| |
Collapse
|
26
|
Xu X, Lu Y, Cao L, Miao Y, Li Y, Cui Y, Han T. Tumor-intrinsic P2RY6 drives immunosuppression by enhancing PGE 2 production. Cell Rep 2024; 43:114469. [PMID: 38996067 DOI: 10.1016/j.celrep.2024.114469] [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: 11/19/2023] [Revised: 05/21/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024] Open
Abstract
Despite the success of anti-programmed cell death-1 (anti-PD-1) immunotherapy, many cancer patients remain unresponsive, and reliable predictive biomarkers are lacking. Here, we show that aberrant expression of the pyrimidinergic receptor P2RY6 is frequent in human cancers and causes immune evasion. In mouse syngeneic and human xenograft tumor models, ectopic expression of P2RY6 shapes an immunosuppressive tumor microenvironment (TME) to enhance tumor growth and resistance to immunotherapy, whereas deletion of P2RY6 from tumors with high P2RY6 expression inflames the TME to inhibit tumor growth. As a G protein-coupled receptor, P2RY6 activates Gq/phospholipase C-β signaling and stimulates the synthesis of prostaglandin E2, which is a key mediator of immunosuppression in the TME. In contrast to the essential role of P2RY6 in tumors, global deletion of P2ry6 from mice does not compromise viability. Our study thus nominates P2RY6 as a precision immunotherapy target for patients with high tumor-intrinsic P2RY6 expression.
Collapse
Affiliation(s)
- Xilong Xu
- College of Life Sciences, Beijing Normal University, Beijing 100875, China; National Institute of Biological Sciences, Beijing 102206, China
| | - Yi Lu
- National Institute of Biological Sciences, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Longzhi Cao
- National Institute of Biological Sciences, Beijing 102206, China; Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yang Miao
- National Institute of Biological Sciences, Beijing 102206, China; PTN Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yamei Li
- National Institute of Biological Sciences, Beijing 102206, China; Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yue Cui
- College of Life Sciences, Beijing Normal University, Beijing 100875, China; National Institute of Biological Sciences, Beijing 102206, China
| | - Ting Han
- College of Life Sciences, Beijing Normal University, Beijing 100875, China; National Institute of Biological Sciences, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China.
| |
Collapse
|
27
|
Farombi EO, Ajayi BO, Ajeigbe OF, Maruf OR, Anyebe DA, Opafunso IT, Adedara IA. Mechanistic exploration of 6-shogaol's preventive effects on azoxymethane and dextran sulfate sodium -induced colorectal cancer: involvement of cell proliferation, apoptosis, carcinoembryonic antigen, wingless-related integration site signaling, and oxido-inflammation. Toxicol Mech Methods 2024:1-10. [PMID: 39034841 DOI: 10.1080/15376516.2024.2381798] [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: 02/01/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
Colorectal cancer (CRC) poses a significant global health burden, being the third most prevalent cancer and the second most significant contributor to cancer-related deaths worldwide. Preventive strategies are crucial to combat this rising incidence. 6-shogaol, derived from ginger, has shown promise in preventing and treating various cancers. This study investigated the preventive effects of 6-shogaol on azoxymethane (AOM) and dextran sulfate sodium (DSS)-induced CRC in mice. Forty male BALB/c mice were randomly divided into control, 6-shogaol, AOM + DSS, and 6-shogaol + AOM + DSS. Mice in the control group received corn oil for 16 weeks, while those in the 6-Shogaol group were administered 20 mg/kg of 6-shogaol for 16 weeks. The AOM + DSS group received a single intraperitoneal dose (ip) of 10 mg/kg of AOM, followed by three cycles of 2.5% DSS in drinking water. The 6-shogaol + AOM + DSS group received both 6-shogaol for 16 weeks and a single ip of 10 mg/kg of AOM, followed by three cycles of 2.5% DSS in drinking water. The AOM + DSS-treated mice exhibited reduced food consumption, colon weight, and colon length, along with increased tumor formation. Co-administration of 6-shogaol effectively reversed these changes, inhibiting CRC development. Histopathological analysis revealed protective effects of 6-shogaol against colonic insults and modulation of inflammatory responses. 6-shogaol significantly reduced Carcinoembryonic antigen and Kiel 67 levels, indicating inhibition of tumor cell proliferation. Mechanistically, 6-shogaol promoted apoptosis by upregulating protein 53 and caspase-3 expression, and it effectively restored the balance of the Wingless-related integration site signaling pathway by regulating β-catenin and adenomatous polyposis coli levels. Moreover, 6-shogaol demonstrated anti-inflammatory effects, reducing myeloperoxidase, Tumor necrosis factor alpha, and cyclooxygenase-2 levels in AOM/DSS-treated mice. Additionally, 6-shogaol restored redox homeostasis by reducing lipid peroxidation and nitrosative stress and enhancing antioxidant enzyme activities. The findings suggest that 6-shogaol inhibits cell proliferation, induces apoptosis, regulates Wnt signaling, suppresses inflammation, and restores redox homeostasis, providing comprehensive insights into its potential therapeutic benefits for CRC.
Collapse
Affiliation(s)
- Ebenezer Olatunde Farombi
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Department of Biochemistry, College of Natural and Applied Sciences, Chrisland University, Abeokuta, Nigeria
| | - Babajide Oluwaseun Ajayi
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Oncopreventives and Systems Oncology Research Laboratory, Biochemistry Unit, Department of Chemical Sciences, Ajayi Crowther University, Oyo, Nigeria
| | - Olufunke Florence Ajeigbe
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Opeyemi Rabiat Maruf
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Daniel Abu Anyebe
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ifeoluwa Tobi Opafunso
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Isaac Adegboyega Adedara
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
28
|
Aradhyula V, Breidenbach JD, Khatib-Shahidi BZ, Slogar JN, Eyong SA, Faleel D, Dube P, Gupta R, Khouri SJ, Haller ST, Kennedy DJ. Transcriptomic Analysis of Arachidonic Acid Pathway Genes Provides Mechanistic Insight into Multi-Organ Inflammatory and Vascular Diseases. Genes (Basel) 2024; 15:954. [PMID: 39062733 PMCID: PMC11275336 DOI: 10.3390/genes15070954] [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: 06/22/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Arachidonic acid (AA) metabolites have been associated with several diseases across various organ systems, including the cardiovascular, pulmonary, and renal systems. Lipid mediators generated from AA oxidation have been studied to control macrophages, T-cells, cytokines, and fibroblasts, and regulate inflammatory mediators that induce vascular remodeling and dysfunction. AA is metabolized by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) to generate anti-inflammatory, pro-inflammatory, and pro-resolutory oxidized lipids. As comorbid states such as diabetes, hypertension, and obesity become more prevalent in cardiovascular disease, studying the expression of AA pathway genes and their association with these diseases can provide unique pathophysiological insights. In addition, the AA pathway of oxidized lipids exhibits diverse functions across different organ systems, where a lipid can be both anti-inflammatory and pro-inflammatory depending on the location of metabolic activity. Therefore, we aimed to characterize the gene expression of these lipid enzymes and receptors throughout multi-organ diseases via a transcriptomic meta-analysis using the Gene Expression Omnibus (GEO) Database. In our study, we found that distinct AA pathways were expressed in various comorbid conditions, especially those with prominent inflammatory risk factors. Comorbidities, such as hypertension, diabetes, and obesity appeared to contribute to elevated expression of pro-inflammatory lipid mediator genes. Our results demonstrate that expression of inflammatory AA pathway genes may potentiate and attenuate disease; therefore, we suggest further exploration of these pathways as therapeutic targets to improve outcomes.
Collapse
Affiliation(s)
- Vaishnavi Aradhyula
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Joshua D. Breidenbach
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Bella Z. Khatib-Shahidi
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Julia N. Slogar
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Sonia A. Eyong
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Dhilhani Faleel
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Prabhatchandra Dube
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Rajesh Gupta
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Samer J. Khouri
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Steven T. Haller
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - David J. Kennedy
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| |
Collapse
|
29
|
Chen Q, Sun Y, Li H. Application of CAR-T cell therapy targeting mesothelin in solid tumor treatment. Discov Oncol 2024; 15:289. [PMID: 39023820 PMCID: PMC11258118 DOI: 10.1007/s12672-024-01159-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy is one of the most effective immunotherapies. CAR-T-cell therapy has achieved great success in the treatment of hematological malignancies. However, due to the characteristics of solid malignant tumors, such as on-target effects, off-tumor toxicity, an immunosuppressive tumor microenvironment (TME), and insufficient trafficking, CAR-T-cell therapy for solid tumors is still in the exploration stage. Mesothelin (MSLN) is a molecule expressed on the surface of various solid malignant tumor cells that is suitable as a target of tumor cells with high MSLN expression for CAR-T-cell therapy. This paper briefly described the development of CAR-T cell therapy and the structural features of MSLN, and especially summarized the strategies of structure optimization of MSLN-targeting CAR-T-cells and the enhancement methods of MSLN-targeting CAR-T cell anti-tumor efficacy by summarizing some preclinical experiment and clinical trials. When considering MSLN-targeting CAR-T-cell therapy as an example, this paper summarizes the efforts made by researchers in CAR-T-cell therapy for solid tumors and summarizes feasible treatment plans by integrating the existing research results.
Collapse
Affiliation(s)
- Qiuhong Chen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, People's Republic of China
| | - Yang Sun
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, People's Republic of China
| | - Hua Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, People's Republic of China.
| |
Collapse
|
30
|
Klekowski J, Chabowski M, Krzystek-Korpacka M, Fleszar M. The Utility of Lipidomic Analysis in Colorectal Cancer Diagnosis and Prognosis-A Systematic Review of Recent Literature. Int J Mol Sci 2024; 25:7722. [PMID: 39062964 PMCID: PMC11277303 DOI: 10.3390/ijms25147722] [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: 05/13/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Colorectal cancer (CRC) is among the most prevalent and lethal malignancies. Lipidomic investigations have revealed numerous disruptions in lipid profiles across various cancers. Studies on CRC exhibit potential for identifying novel diagnostic or prognostic indicators through lipidomic signatures. This review examines recent literature regarding lipidomic markers for CRC. PubMed database was searched for eligible articles concerning lipidomic biomarkers of CRC. After selection, 36 articles were included in the review. Several studies endeavor to establish sets of lipid biomarkers that demonstrate promising potential to diagnose CRC based on blood samples. Phosphatidylcholine, phosphatidylethanolamine, ceramides, and triacylglycerols (TAGs) appear to offer the highest diagnostic accuracy. In tissues, lysophospholipids, ceramides, and TAGs were among the most altered lipids, while unsaturated fatty acids also emerged as potential biomarkers. In-depth analysis requires both cell culture and animal studies. CRC involves multiple lipid metabolism alterations. Although numerous lipid species have been suggested as potential diagnostic markers, the establishment of standardized methods and the conduct of large-scale studies are necessary to facilitate their clinical application.
Collapse
Affiliation(s)
- Jakub Klekowski
- Department of Nursing and Obstetrics, Division of Anesthesiological and Surgical Nursing, Faculty of Health Science, Wroclaw Medical University, 50-367 Wroclaw, Poland;
- Department of Surgery, 4th Military Clinical Hospital, 50-981 Wroclaw, Poland
| | - Mariusz Chabowski
- Department of Surgery, 4th Military Clinical Hospital, 50-981 Wroclaw, Poland
- Department of Clinical Surgical Sciences, Faculty of Medicine, Wroclaw University of Science and Technology, 50-556 Wroclaw, Poland
| | - Małgorzata Krzystek-Korpacka
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.K.-K.); (M.F.)
| | - Mariusz Fleszar
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.K.-K.); (M.F.)
- Omics Research Center, Wroclaw Medical University, 50-368 Wroclaw, Poland
| |
Collapse
|
31
|
Beton-Mysur K, Surmacki J, Brożek-Płuska B. Raman-AFM-fluorescence-guided impact of linoleic and eicosapentaenoic acids on subcellular structure and chemical composition of normal and cancer human colon cells. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 315:124242. [PMID: 38581725 DOI: 10.1016/j.saa.2024.124242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/28/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024]
Abstract
The regular overconsumption of high-energy food (rich in lipids and sugars) results in elevated nutrient absorption in intestine and consequently excessive accumulation of lipids in many organs e.g.: liver, adipose tissue, muscles. In the long term this can lead to obesity and obesity-associated diseases e.g. type 2 diabetes, non-alcoholic fatty liver disease, cardiovascular disease, inflammatory bowel disease (IBD). In the presented paper based on RI data we have proved that Raman maps can be used successfully for subcellular structures visualization and analysis of fatty acids impact on morphology and chemical composition of human colon single cells - normal and cancer. Based on Raman data we have investigated the changes related to endoplasmic reticulum, mitochondria, lipid droplets and nucleus. Analysis of ratios calculated based on Raman bands typical for proteins (1256, 1656 cm-1), lipids (1304, 1444 cm-1) and nucleic acids (750 cm-1) has confirmed for endoplasmic reticulum the increased activity of this organelle in lipoproteins synthesis upon FAs supplementation; for LDs the changes of desaturation of accumulated lipids with the highest unsaturation level for CaCo-2 cells upon EPA supplementation; for mitochondria the stronger effect of FAs supplementation was observed for CaCo-2 cells confirming the increased activity of this organelle responsible for energy production necessary for tumor development; the weakest impact of FAs supplementation was observed for nucleus for both types of cells and both types of acids. Fluorescence imaging was used for the investigations of changes in LDs/ER morphology. Our measurements have shown the increased area of LDs/ER for CaCo-2 cancer cells, and the strongest effect was noticed for CaCo-2 cells upon EPA supplementation. The increased participation of lipid structures for all types of cells upon FAs supplementation has been confirmed also by AFM studies. The lowest YM values have been observed for CaCo-2 cells including samples treated with FAs.
Collapse
Affiliation(s)
- Karolina Beton-Mysur
- Lodz University of Technology, Faculty of Chemistry, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, Wroblewskiego 15, 93-590 Lodz, Poland
| | - Jakub Surmacki
- Lodz University of Technology, Faculty of Chemistry, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, Wroblewskiego 15, 93-590 Lodz, Poland
| | - Beata Brożek-Płuska
- Lodz University of Technology, Faculty of Chemistry, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, Wroblewskiego 15, 93-590 Lodz, Poland.
| |
Collapse
|
32
|
Xue Y, Chen Y, Sun S, Tong X, Chen Y, Tang S, Wang X, Bi S, Qiu Y, Zhao Q, Qin Z, Xu Q, Ai Y, Chen L, Zhang B, Liu Z, Ji M, Lang M, Chen L, Xu G, Hu L, Ye D, Ji H. TET2-STAT3-CXCL5 nexus promotes neutrophil lipid transfer to fuel lung adeno-to-squamous transition. J Exp Med 2024; 221:e20240111. [PMID: 38805014 PMCID: PMC11129275 DOI: 10.1084/jem.20240111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/21/2024] [Accepted: 04/05/2024] [Indexed: 05/29/2024] Open
Abstract
Phenotypic plasticity is a rising cancer hallmark, and lung adeno-to-squamous transition (AST) triggered by LKB1 inactivation is significantly associated with drug resistance. Mechanistic insights into AST are urgently needed to identify therapeutic vulnerability in LKB1-deficient lung cancer. Here, we find that ten-eleven translocation (TET)-mediated DNA demethylation is elevated during AST in KrasLSL-G12D/+; Lkb1L/L (KL) mice, and knockout of individual Tet genes reveals that Tet2 is required for squamous transition. TET2 promotes neutrophil infiltration through STAT3-mediated CXCL5 expression. Targeting the STAT3-CXCL5 nexus effectively inhibits squamous transition through reducing neutrophil infiltration. Interestingly, tumor-infiltrating neutrophils are laden with triglycerides and can transfer the lipid to tumor cells to promote cell proliferation and squamous transition. Pharmacological inhibition of macropinocytosis dramatically inhibits neutrophil-to-cancer cell lipid transfer and blocks squamous transition. These data uncover an epigenetic mechanism orchestrating phenotypic plasticity through regulating immune microenvironment and metabolic communication, and identify therapeutic strategies to inhibit AST.
Collapse
Affiliation(s)
- Yun Xue
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuting Chen
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Sijia Sun
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), and Molecular and Cell Biology Laboratory, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xinyuan Tong
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yujia Chen
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), and Molecular and Cell Biology Laboratory, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Shijie Tang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xue Wang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Simin Bi
- Department of Physics, State Key Laboratory of Surface Physics, Academy for Engineering and Technology, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Shanghai, China
| | - Yuqin Qiu
- Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Qiqi Zhao
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Zhen Qin
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Qin Xu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yingjie Ai
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Leilei Chen
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), and Molecular and Cell Biology Laboratory, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Beizhen Zhang
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhijie Liu
- Department of Physics, State Key Laboratory of Surface Physics, Academy for Engineering and Technology, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Shanghai, China
| | - Minbiao Ji
- Department of Physics, State Key Laboratory of Surface Physics, Academy for Engineering and Technology, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Shanghai, China
| | - Meidong Lang
- Key Laboratory of Advanced Polymeric Materials, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
- Department of General Surgery and Laboratory of General Surgery, Xinhua Hospital, Affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Luonan Chen
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Guoliang Xu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Chinese Academy of Medical Sciences (RU069), Shanghai, China
| | - Liang Hu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Dan Ye
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), and Molecular and Cell Biology Laboratory, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Hongbin Ji
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| |
Collapse
|
33
|
Yang J, Li Y, Han X, Li T, Li D, Liu Q, Yan L, Li F, Pei X, Feng Y, Lin Z, Fu Z, Wang C, Sun Q, Li C. Targeting estrogen mediated CYP4F2/CYP4F11-20-HETE metabolic disorder decelerates tumorigenesis in ER+ breast cancer. Biochem Biophys Rep 2024; 38:101706. [PMID: 38646426 PMCID: PMC11033080 DOI: 10.1016/j.bbrep.2024.101706] [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: 09/24/2023] [Revised: 03/25/2024] [Accepted: 04/01/2024] [Indexed: 04/23/2024] Open
Abstract
Purpose As the most common subset of breast cancer (BC), estrogen receptor positive (ER+) BC accounting for 80% of cases, has become a global public health concern. The female hormone estrogen (E2) unequivocally drives ER + breast malignancies. The reasons that estrogen affects BC development has long been considered, yet further study remains to be conducted of the molecular events in the E2-estrogen receptor α (ERα) signaling pathway in ER + BC progression, especially lipid metabolism, so providing more options for tailored and individualized therapy. Our aim is to find out new targets and clinical biomarkers for ER + breast cancer treatment from the perspective of lipid metabolism. Methods Lipid metabolomics profiling was used to examine the membrane phospholipid stimulated by E2. Clinical BC samples were used to assess the association of CYP4F2, CYP4F11 expression with clinicopathological characteristics and patient outcomes. Some inhibitors of main enzymes in AA metabolism were used combined with E2 to assess roles of CYP4F2/CYP4F11 in the progression of ER + BC. CYP4F2, CYP4F11 overexpression and knockdown BC cell lines were employed to examine the effects of CYP4F2, CYP4F11 on cellular proliferation, apoptosis and tumor growth. Western blotting, qPCR, Immunohistochemical staining and flow cytometry were also conducted to determine the underlying mechanisms related to CYP4F2, CYP4F11 function. Results The activation of the CYP450 signaling pathway in arachidonic acid metabolism contributed to ER + BC tumorigenesis. In ER + BC, CYP4F2 and CYP4F11 overexpression induced by E2 could promote cancer cell proliferation and resistance to apoptosis by producing the metabolite 20-HETE and activating the antiapoptotic protein Bcl-2. CYP4F2 and CYP4F11 elevation correlates with poorer overall survival and disease-free survival in ER + BC patients. Conclusion CYP4F2, CYP4F11 and their metabolite 20-HETE could serve as effective prognostic markers and attractive therapeutic targets for novel anticancer drug development about ER + BC.
Collapse
Affiliation(s)
- Juan Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, PR China
| | - Yin Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, PR China
| | - Xiao Han
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, PR China
| | - Tianjiao Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, PR China
| | - Ding Li
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Qiao Liu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, PR China
| | - Lizhong Yan
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, PR China
| | - Fei Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, PR China
| | - Xiaolin Pei
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, PR China
| | - Ya Feng
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, PR China
| | - Zhoujun Lin
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, PR China
| | - Zhenkun Fu
- Department of Immunology & Wu Lien-Teh Institute & Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin Medical University & Heilongjiang Academy of Medical Science, Harbin, 150081, PR China
| | - Changjun Wang
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing, 100730, PR China
| | - Qiang Sun
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing, 100730, PR China
| | - Chenggang Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300350, PR China
| |
Collapse
|
34
|
Lorito N, Subbiani A, Smiriglia A, Bacci M, Bonechi F, Tronci L, Romano E, Corrado A, Longo DL, Iozzo M, Ippolito L, Comito G, Giannoni E, Meattini I, Avgustinova A, Chiarugi P, Bachi A, Morandi A. FADS1/2 control lipid metabolism and ferroptosis susceptibility in triple-negative breast cancer. EMBO Mol Med 2024; 16:1533-1559. [PMID: 38926633 PMCID: PMC11251055 DOI: 10.1038/s44321-024-00090-6] [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: 06/29/2023] [Revised: 05/24/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Triple-negative breast cancer (TNBC) has limited therapeutic options, is highly metastatic and characterized by early recurrence. Lipid metabolism is generally deregulated in TNBC and might reveal vulnerabilities to be targeted or used as biomarkers with clinical value. Ferroptosis is a type of cell death caused by iron-dependent lipid peroxidation which is facilitated by the presence of polyunsaturated fatty acids (PUFA). Here we identify fatty acid desaturases 1 and 2 (FADS1/2), which are responsible for PUFA biosynthesis, to be highly expressed in a subset of TNBC with a poorer prognosis. Lipidomic analysis, coupled with functional metabolic assays, showed that FADS1/2 high-expressing TNBC are susceptible to ferroptosis-inducing agents and that targeting FADS1/2 by both genetic interference and pharmacological approach renders those tumors ferroptosis-resistant while unbalancing PUFA/MUFA ratio by the supplementation of exogenous PUFA sensitizes resistant tumors to ferroptosis induction. Last, inhibiting lipid droplet (LD) formation and turnover suppresses the buffering capacity of LD and potentiates iron-dependent cell death. These findings have been validated in vitro and in vivo in mouse- and human-derived clinically relevant models and in a retrospective cohort of TNBC patients.
Collapse
Affiliation(s)
- Nicla Lorito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Angela Subbiani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Alfredo Smiriglia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Marina Bacci
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Francesca Bonechi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Laura Tronci
- IFOM ETS - The AIRC Institute of Molecular Oncology, Via Adamello 16, 20139, Milan, Italy
| | - Elisabetta Romano
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Alessia Corrado
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126, Torino, Italy
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), National Research Council of Italy (CNR), Via Nizza 52, 10126, Torino, Italy
| | - Marta Iozzo
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Luigi Ippolito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Giuseppina Comito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Icro Meattini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy
| | - Alexandra Avgustinova
- Institut de Recerca Sant Joan de Déu, Carrer Santa Rosa 39-57, 08950, Esplugues de Llobregat, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Angela Bachi
- IFOM ETS - The AIRC Institute of Molecular Oncology, Via Adamello 16, 20139, Milan, Italy
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy.
| |
Collapse
|
35
|
Maiuolo J, Bulotta RM, Ruga S, Nucera S, Macrì R, Scarano F, Oppedisano F, Carresi C, Gliozzi M, Musolino V, Mollace R, Muscoli C, Mollace V. The Postbiotic Properties of Butyrate in the Modulation of the Gut Microbiota: The Potential of Its Combination with Polyphenols and Dietary Fibers. Int J Mol Sci 2024; 25:6971. [PMID: 39000076 PMCID: PMC11240906 DOI: 10.3390/ijms25136971] [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: 05/28/2024] [Revised: 06/15/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024] Open
Abstract
The gut microbiota is a diverse bacterial community consisting of approximately 2000 species, predominantly from five phyla: Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, and Verrucomicrobia. The microbiota's bacterial species create distinct compounds that impact the host's health, including well-known short-chain fatty acids. These are produced through the breakdown of dietary fibers and fermentation of undigested carbohydrates by the intestinal microbiota. The main short-chain fatty acids consist of acetate, propionate, and butyrate. The concentration of butyrate in mammalian intestines varies depending on the diet. Its main functions are use as an energy source, cell differentiation, reduction in the inflammatory process in the intestine, and defense against oxidative stress. It also plays an epigenetic role in histone deacetylases, thus helping to reduce the risk of colon cancer. Finally, butyrate affects the gut-brain axis by crossing the brain-blood barrier, making it crucial to determine the right concentrations for both local and peripheral effects. In recent years, there has been a significant amount of attention given to the role of dietary polyphenols and fibers in promoting human health. Polyphenols and dietary fibers both play crucial roles in protecting human health and can produce butyrate through gut microbiota fermentation. This paper aims to summarize information on the key summits related to the negative correlation between intestinal microbiota diversity and chronic diseases to guide future research on determining the specific activity of butyrate from polyphenols and dietary fibers that can carry out these vital functions.
Collapse
Affiliation(s)
- Jessica Maiuolo
- IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (R.M.B.); (S.R.); (S.N.); (R.M.); (F.S.); (F.O.); (C.C.); (M.G.); (V.M.); (C.M.); (V.M.)
| | - Rosa Maria Bulotta
- IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (R.M.B.); (S.R.); (S.N.); (R.M.); (F.S.); (F.O.); (C.C.); (M.G.); (V.M.); (C.M.); (V.M.)
| | - Stefano Ruga
- IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (R.M.B.); (S.R.); (S.N.); (R.M.); (F.S.); (F.O.); (C.C.); (M.G.); (V.M.); (C.M.); (V.M.)
| | - Saverio Nucera
- IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (R.M.B.); (S.R.); (S.N.); (R.M.); (F.S.); (F.O.); (C.C.); (M.G.); (V.M.); (C.M.); (V.M.)
| | - Roberta Macrì
- IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (R.M.B.); (S.R.); (S.N.); (R.M.); (F.S.); (F.O.); (C.C.); (M.G.); (V.M.); (C.M.); (V.M.)
| | - Federica Scarano
- IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (R.M.B.); (S.R.); (S.N.); (R.M.); (F.S.); (F.O.); (C.C.); (M.G.); (V.M.); (C.M.); (V.M.)
| | - Francesca Oppedisano
- IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (R.M.B.); (S.R.); (S.N.); (R.M.); (F.S.); (F.O.); (C.C.); (M.G.); (V.M.); (C.M.); (V.M.)
| | - Cristina Carresi
- IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (R.M.B.); (S.R.); (S.N.); (R.M.); (F.S.); (F.O.); (C.C.); (M.G.); (V.M.); (C.M.); (V.M.)
| | - Micaela Gliozzi
- IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (R.M.B.); (S.R.); (S.N.); (R.M.); (F.S.); (F.O.); (C.C.); (M.G.); (V.M.); (C.M.); (V.M.)
| | - Vincenzo Musolino
- IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (R.M.B.); (S.R.); (S.N.); (R.M.); (F.S.); (F.O.); (C.C.); (M.G.); (V.M.); (C.M.); (V.M.)
| | - Rocco Mollace
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Roma, Italy;
| | - Carolina Muscoli
- IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (R.M.B.); (S.R.); (S.N.); (R.M.); (F.S.); (F.O.); (C.C.); (M.G.); (V.M.); (C.M.); (V.M.)
| | - Vincenzo Mollace
- IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (R.M.B.); (S.R.); (S.N.); (R.M.); (F.S.); (F.O.); (C.C.); (M.G.); (V.M.); (C.M.); (V.M.)
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Roma, Italy;
| |
Collapse
|
36
|
Zhang T, Zeng X, Zeng E, Wang H. Ferroptosis in antitumor therapy: Unraveling regulatory mechanisms and immunogenic potential. Int Immunopharmacol 2024; 134:112203. [PMID: 38705030 DOI: 10.1016/j.intimp.2024.112203] [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: 02/12/2024] [Revised: 04/17/2024] [Accepted: 05/01/2024] [Indexed: 05/07/2024]
Abstract
Ferroptosis, a recently discovered form of non-apoptotic cell death, has the potential to revolutionize anti-tumor therapy. This review highlights the regulatory mechanisms and immunogenic properties of ferroptosis, and how it can enhance the effectiveness of radio and immunotherapies in overcoming tumor resistance. However, tumor metabolism and the impact of ferroptosis on the tumor microenvironment present challenges in completely realizing its therapeutic potential. A deeper understanding of the effects of ferroptosis on tumor cells and their associated immune cells is essential for developing more effective tumor treatment strategies. This review offers a comprehensive overview of the relationship between ferroptosis and tumor immunity, and sheds new light on its application in tumor immunotherapy.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China; First Clinical Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Xiaoping Zeng
- Medical College, Jinhua Polytechnic, Jinhua 321017, Zhejiang Province, China; School of Basic Medical Sciences, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Erming Zeng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China.
| | - Hongmei Wang
- Medical College, Jinhua Polytechnic, Jinhua 321017, Zhejiang Province, China; School of Basic Medical Sciences, Nanchang University, Nanchang 330006, Jiangxi Province, China.
| |
Collapse
|
37
|
Sipos F, Műzes G. Colonic Tuft Cells: The Less-Recognized Therapeutic Targets in Inflammatory Bowel Disease and Colorectal Cancer. Int J Mol Sci 2024; 25:6209. [PMID: 38892399 PMCID: PMC11172904 DOI: 10.3390/ijms25116209] [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: 04/28/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Tuft cells are more than guardian chemosensory elements of the digestive tract. They produce a variety of immunological effector molecules in response to stimulation; moreover, they are essential for defense against protozoa and nematodes. Beyond the description of their characteristics, this review aims to elucidate the potential pathogenic and therapeutic roles of colonic tuft cells in inflammatory bowel disease and colorectal cancer, focusing on their primarily immunomodulatory action. Regarding inflammatory bowel disease, tuft cells are implicated in both maintaining the integrity of the intestinal epithelial barrier and in tissue repair and regeneration processes. In addition to maintaining intestinal homeostasis, they display complex immune-regulatory functions. During the development of colorectal cancer, tuft cells can promote the epithelial-to-mesenchymal transition, alter the gastrointestinal microenvironment, and modulate both the anti-tumor immune response and the tumor microenvironment. A wide variety of their biological functions can be targeted for anti-inflammatory or anti-tumor therapies; however, the adverse side effects of immunomodulatory actions must be strictly considered.
Collapse
Affiliation(s)
- Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| | - Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
38
|
Bi X, Wang Y, Lin Y, Wang M, Li X. Genetic Evidence for Causal Relationships between Plasma Eicosanoid Levels and Cardiovascular Disease. Metabolites 2024; 14:294. [PMID: 38921429 PMCID: PMC11206149 DOI: 10.3390/metabo14060294] [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: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Cardiovascular diseases are the most common causes of mortality and disability worldwide. Eicosanoids are a group of bioactive metabolites that are mainly oxidized by arachidonic acid. Eicosanoids play a diverse role in cardiovascular diseases, with some exerting beneficial effects while others have detrimental consequences. However, a causal relationship between eicosanoid levels and cardiovascular disease remains unclear. Six single nucleotide polymorphisms (SNPs) with strong associations with plasma eicosanoid levels were selected. Summary-level data for cardiovascular disease were obtained from publicly available genome-wide association studies. A two-sample MR analysis identified that plasma eicosanoid levels were inversely correlated with unstable angina pectoris (OR 1.06; 95% CI 1-1.12; p = 0.04), myocardial infarction (OR 1.05; 95% CI 1.02-1.09; p = 0.005), ischemia stroke (OR 1.05; 95% CI 1-1.11; p = 0.047), transient ischemic attack (OR 1.03; 95% CI 1-1.07; p = 0.042), heart failure (OR 1.03; 95% CI 1.01-1.05; p = 0.011), and pulmonary embolism (OR 1.08; 95% CI 1.02-1.14; p = 1.69 × 10-6). In conclusion, our data strongly suggest a genetic causal link between high plasma eicosanoid levels and an increased cardiovascular disease risk. This study provides genetic evidence for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Xukun Bi
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yiran Wang
- Department of Nursing, No. 906 Hospital of People’s Liberation Army, Ningbo 315000, China
| | - Yangjun Lin
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Meihui Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Xiaoting Li
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| |
Collapse
|
39
|
Kowalski JP, Rettie AE. There and Back Again: A Perspective on 20 Years of CYP4Z1. Drug Metab Dispos 2024; 52:498-507. [PMID: 38604728 DOI: 10.1124/dmd.124.001670] [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: 01/29/2024] [Revised: 03/17/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
Cytochrome P450 (CYP)4Z1, a highly expressed CYP gene in breast cancer, was one of the last CYPs to be identified in the human genome, some 20 years ago. CYP4 enzymes typically catalyze ω-hydroxylation and metabolize ω3 and ω6 polyunsaturated fatty acids to bioactive lipid metabolites that can influence tumor growth and metastasis. These attributes of CYP4Z1 make it an attractive target for new chemotherapeutic drug design, as a potential biomarker for selection of patients that might respond favorably to drugs and for developing enzyme inhibitors as potential therapeutic agents. This review summarizes the current state of knowledge regarding the advancing biochemistry of CYP4Z1, its role in breast cancer, and the recent synthesis of selective chemical inhibitors of the enzyme. We identify gaps that need to be filled to further advance this field and present new experimental data on recombinant CYP4Z1 expression and purification of the active catalytic form. SIGNIFICANCE STATEMENT: In breast cancer, an unmet need is the availability of highly effective therapeutic agents, especially for triple negative breast cancer. The relevance of the work summarized in this mini-review is that it identifies a new potential drug target, CYP4Z1, and discusses ways in which the gene product's catalytic activity might be modulated in order to combat this malignancy and limit its spread.
Collapse
Affiliation(s)
- John P Kowalski
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington
| | - Allan E Rettie
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington
| |
Collapse
|
40
|
Kudo K, Yanagiya R, Hasegawa M, Carreras J, Miki Y, Nakayama S, Nagashima E, Miyatake Y, Torii K, Ando K, Nakamura N, Miyajima A, Murakami M, Kotani A. Unique lipid composition maintained by extracellular blockade leads to prooncogenicity. Cell Death Discov 2024; 10:221. [PMID: 38719806 PMCID: PMC11079073 DOI: 10.1038/s41420-024-01971-y] [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: 12/21/2023] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
Lipid-mediated inflammation is involved in the development and malignancy of cancer. We previously demonstrated the existence of a novel oncogenic mechanism utilizing membrane lipids of extracellular vesicles in Epstein-Barr virus (EBV)-positive lymphomas and found that the lipid composition of lymphoma cells is skewed toward ω-3 fatty acids, which are anti-inflammatory lipids, suggesting an alteration in systemic lipid composition. The results showed that arachidonic acid (AA), an inflammatory lipid, was significantly reduced in the infected cells but detected at high levels in the sera of EBV-positive patients lead to the finding of the blockade of extracellular AA influx by downregulating FATP2, a long-chain fatty acid transporter that mainly transports AA in EBV-infected lymphoma cells. Low AA levels in tumor cells induced by downregulation of FATP2 expression confer resistance to ferroptosis and support tumor growth. TCGA data analysis and xenograft models have demonstrated that the axis plays a critical role in several types of cancers, especially poor prognostic cancers, such as glioblastoma and melanoma. Overall, our in vitro, in vivo, in silico, and clinical data suggest that several cancers exert oncogenic activity by maintaining their special lipid composition via extracellular blockade.
Collapse
Affiliation(s)
- Kai Kudo
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, Japan
| | - Ryo Yanagiya
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Department of Hematology and Oncology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Laboratory of Regulation of Infectious Cancer, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Masanori Hasegawa
- Department of Urology, Tokai University School of Medicine, Isehara, Kanagawa, Japan.
| | - Joaquim Carreras
- Department of Pathology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yoshimi Miki
- Laboratory of Microenvironmental Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shunya Nakayama
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, Japan
- Laboratory of Veterinary Physiology, College of Bioresource Science, Nihon University, Fujisawa, Kanagawa, Japan
| | - Etsuko Nagashima
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, Japan
| | - Yuji Miyatake
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, Japan
| | - Kan Torii
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Kiyoshi Ando
- Department of Hematology and Oncology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Naoya Nakamura
- Department of Pathology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Akira Miyajima
- Department of Urology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Makoto Murakami
- Laboratory of Microenvironmental Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ai Kotani
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan.
- Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, Japan.
- Laboratory of Regulation of Infectious Cancer, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
41
|
Gilad O, Muller C, Kupfer SS. Chemoprevention in Inherited Colorectal Cancer Syndromes. Clin Colon Rectal Surg 2024; 37:172-179. [PMID: 38606042 PMCID: PMC11006448 DOI: 10.1055/s-0043-1770384] [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] [Indexed: 04/13/2024]
Abstract
Cancer prevention in hereditary gastrointestinal predisposition syndromes relies primarily on intensive screening (e.g., colonoscopy) or prophylactic surgery (e.g., colectomy). The use of chemopreventive agents as an adjunct to these measures has long been studied both in the general population and in hereditary cancer patients, in whom the risk of malignancy, and therefore the potential risk reduction, is considerably greater. However, to date only few compounds have been found to be effective, safe, and tolerable for widespread use. Furthermore, many of the studies involving these rare syndromes suffer from small sample sizes, heterogeneous patient cohorts, short follow-up duration, and lack of standardized endpoints, creating challenges to draw generalizable conclusion regarding efficacy. The following review summarizes the current data on various chemopreventive compounds used in Lynch syndrome and familial adenomatous polyposis in addition to several agents that are currently being investigated.
Collapse
Affiliation(s)
- Ophir Gilad
- Section of Gastroenterology, Hepatology and Nutrition, University of Chicago, Chicago, Illinois
| | - Charles Muller
- Division of Gastroenterology and Hepatology, Northwestern University, Chicago, Illinois
| | - Sonia S. Kupfer
- Section of Gastroenterology, Hepatology and Nutrition, University of Chicago, Chicago, Illinois
| |
Collapse
|
42
|
Chen Y, Zhong Z, Deng Y, Lu Y, Qin X. M2 tumor-associated macrophages and CXCL2 induce lipid remodeling in hepatocellular carcinoma cell lines. Biomed Chromatogr 2024; 38:e5837. [PMID: 38316604 DOI: 10.1002/bmc.5837] [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: 10/17/2023] [Revised: 12/09/2023] [Accepted: 01/11/2024] [Indexed: 02/07/2024]
Abstract
Primary hepatocellular carcinoma (HCC) is one of the most common malignant tumors, but its pathogenesis remains incompletely elucidated. Recently, many studies indicated that lipid remodeling plays an important role in the occurrence and development of HCC. Furthermore, lipids have been proven to be indispensable mediators in promoting communication between tumor cells and extracellular matrix in the tumor microenvironment. Thus, this study aims to comprehensively investigate the process of lipid remodeling during HCC metastasis based on the LC-electrospray ionization-MS (LC-ESI-MS) combined with multiple reaction monitoring technology. M2 tumor-associated macrophages and the recombinant human protein CXCL2 were used to simulate the tumor microenvironment. After co-incubating SMMC7721 and MHCC97-H cell lines with M2 tumor-associated macrophages or the recombinant human protein CXCL2 for 48 h, LC-ESI-MS was used to quantify the levels of two major classes of lipid molecules, namely, glycerophospholipids and sphingolipids. Our results suggest that lipid remodeling in the tumor microenvironment may promote the migration and invasion of HCC cell lines.
Collapse
Affiliation(s)
- Yongling Chen
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ziqing Zhong
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yan Deng
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yu Lu
- Department of Laboratory Medicine, Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
| | - Xue Qin
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
43
|
Lacher SB, Dörr J, de Almeida GP, Hönninger J, Bayerl F, Hirschberger A, Pedde AM, Meiser P, Ramsauer L, Rudolph TJ, Spranger N, Morotti M, Grimm AJ, Jarosch S, Oner A, Gregor L, Lesch S, Michaelides S, Fertig L, Briukhovetska D, Majed L, Stock S, Busch DH, Buchholz VR, Knolle PA, Zehn D, Dangaj Laniti D, Kobold S, Böttcher JP. PGE 2 limits effector expansion of tumour-infiltrating stem-like CD8 + T cells. Nature 2024; 629:417-425. [PMID: 38658748 PMCID: PMC11078747 DOI: 10.1038/s41586-024-07254-x] [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: 05/22/2023] [Accepted: 02/28/2024] [Indexed: 04/26/2024]
Abstract
Cancer-specific TCF1+ stem-like CD8+ T cells can drive protective anticancer immunity through expansion and effector cell differentiation1-4; however, this response is dysfunctional in tumours. Current cancer immunotherapies2,5-9 can promote anticancer responses through TCF1+ stem-like CD8+ T cells in some but not all patients. This variation points towards currently ill-defined mechanisms that limit TCF1+CD8+ T cell-mediated anticancer immunity. Here we demonstrate that tumour-derived prostaglandin E2 (PGE2) restricts the proliferative expansion and effector differentiation of TCF1+CD8+ T cells within tumours, which promotes cancer immune escape. PGE2 does not affect the priming of TCF1+CD8+ T cells in draining lymph nodes. PGE2 acts through EP2 and EP4 (EP2/EP4) receptor signalling in CD8+ T cells to limit the intratumoural generation of early and late effector T cell populations that originate from TCF1+ tumour-infiltrating CD8+ T lymphocytes (TILs). Ablation of EP2/EP4 signalling in cancer-specific CD8+ T cells rescues their expansion and effector differentiation within tumours and leads to tumour elimination in multiple mouse cancer models. Mechanistically, suppression of the interleukin-2 (IL-2) signalling pathway underlies the PGE2-mediated inhibition of TCF1+ TIL responses. Altogether, we uncover a key mechanism that restricts the IL-2 responsiveness of TCF1+ TILs and prevents anticancer T cell responses that originate from these cells. This study identifies the PGE2-EP2/EP4 axis as a molecular target to restore IL-2 responsiveness in anticancer TILs to achieve cancer immune control.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Differentiation
- Cell Line, Tumor
- Cell Proliferation
- Dinoprostone/metabolism
- Disease Models, Animal
- Hepatocyte Nuclear Factor 1-alpha/metabolism
- Interleukin-2
- Lymph Nodes/cytology
- Lymph Nodes/immunology
- Lymphocytes, Tumor-Infiltrating/cytology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mice, Inbred C57BL
- Neoplasms/immunology
- Neoplasms/prevention & control
- Receptors, Prostaglandin E, EP2 Subtype/deficiency
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/deficiency
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Signal Transduction
- Stem Cells/cytology
- Stem Cells/immunology
- Stem Cells/metabolism
- Tumor Escape/immunology
Collapse
Affiliation(s)
- Sebastian B Lacher
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Janina Dörr
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Member of the German Center for Lung Research (DZL), LMU Munich, Munich, Germany
| | - Gustavo P de Almeida
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, TUM, Freising, Germany
| | - Julian Hönninger
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
- Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, TUM, Munich, Germany
| | - Felix Bayerl
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Anna Hirschberger
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Anna-Marie Pedde
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Philippa Meiser
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Lukas Ramsauer
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Thomas J Rudolph
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Nadine Spranger
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Matteo Morotti
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, University Hospital of Lausanne (CHUV) and UNIL, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Alizee J Grimm
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, University Hospital of Lausanne (CHUV) and UNIL, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Sebastian Jarosch
- Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, TUM, Munich, Germany
- Boehringer Ingelheim, Biberach, Germany
| | - Arman Oner
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Member of the German Center for Lung Research (DZL), LMU Munich, Munich, Germany
| | - Lisa Gregor
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Member of the German Center for Lung Research (DZL), LMU Munich, Munich, Germany
| | - Stefanie Lesch
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Member of the German Center for Lung Research (DZL), LMU Munich, Munich, Germany
| | - Stefanos Michaelides
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Member of the German Center for Lung Research (DZL), LMU Munich, Munich, Germany
| | - Luisa Fertig
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Member of the German Center for Lung Research (DZL), LMU Munich, Munich, Germany
| | - Daria Briukhovetska
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Member of the German Center for Lung Research (DZL), LMU Munich, Munich, Germany
| | - Lina Majed
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Member of the German Center for Lung Research (DZL), LMU Munich, Munich, Germany
| | - Sophia Stock
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Member of the German Center for Lung Research (DZL), LMU Munich, Munich, Germany
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, TUM, Munich, Germany
| | - Veit R Buchholz
- Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine and Health, TUM, Munich, Germany
| | - Percy A Knolle
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, TUM, Freising, Germany
| | - Denarda Dangaj Laniti
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, University Hospital of Lausanne (CHUV) and UNIL, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, Member of the German Center for Lung Research (DZL), LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Munich, Research Center for Environmental Health (HMGU), Neuherberg, Germany
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany.
| |
Collapse
|
44
|
Yamada J, Fukui T, Yatani A, Mimura C, Fukuda K, Hazama D, Katsurada N, Nagano T, Yamamoto M, Tachihara M. Impact of concurrent medications on the outcome of immunotherapy in non-small cell lung carcinoma. Thorac Cancer 2024; 15:1228-1236. [PMID: 38622898 PMCID: PMC11128370 DOI: 10.1111/1759-7714.15294] [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: 01/25/2024] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND There have been reports on the impact of concurrent drugs on the outcome of immunotherapy for non-small cell lung carcinoma (NSCLC). However, the effect of some drugs, such as antibiotics and nonsteroidal anti-inflammatory drugs (NSAIDs), has not been clarified in patients with NSCLC. In the present study, we aimed to assess the association between concurrent drugs and the outcomes of immune checkpoint inhibitors (ICIs) alone or in combination with chemotherapy for patients with advanced NSCLC. METHODS We retrospectively assessed patients with advanced NSCLC who underwent ICI treatment between September 2017 and December 2021 at Kobe University Hospital. We evaluated the data regarding the use of antibiotics within 30 days before ICI initiation, as well as the use of proton pump inhibitors (PPIs) and NSAIDs during ICI initiation. RESULTS A total of 127 patients were assessed, among whom 28 (22.0%) patients received antibiotics, 39 (30.7%) PPIs, and 36 (28.3%) NSAIDs. No significant differences were observed between the patients with and without antibiotic use. However, patients using NSAIDs had significantly worse objective response rates (ORR) and progression-free survival (PFS) with ICI alone or in combination with chemotherapy compared to those who did not (ORR, 47.2% vs. 67.0%; p = 0.045. PFS, 6.3 months vs. 10.8 months; p = 0.02). Patients using PPIs demonstrated a worse ORR of ICI in combination with chemotherapy compared to those who did not (ORR, 45.2% vs. 72.6%; p = 0.013). CONCLUSIONS The unnecessary use of NSAIDs along with immunotherapy should be discouraged.
Collapse
Affiliation(s)
- Jun Yamada
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Takafumi Fukui
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Atsuhiko Yatani
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Chihiro Mimura
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Kiyoko Fukuda
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Daisuke Hazama
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Naoko Katsurada
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Masatsugu Yamamoto
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Motoko Tachihara
- Division of Respiratory Medicine, Department of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| |
Collapse
|
45
|
Murray M. Omega-3 polyunsaturated fatty acid derived lipid mediators: a comprehensive update on their application in anti-cancer drug discovery. Expert Opin Drug Discov 2024; 19:617-629. [PMID: 38595031 DOI: 10.1080/17460441.2024.2340493] [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: 02/26/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
INTRODUCTION ω-3 Polyunsaturated fatty acids (PUFAs) have a range of health benefits, including anticancer activity, and are converted to lipid mediators that could be adapted into pharmacological strategies. However, the stability of these mediators must be improved, and they may require formulation to achieve optimal tissue concentrations. AREAS COVERED Herein, the author reviews the literature around chemical stabilization and formulation of ω-3 PUFA mediators and their application in anticancer drug discovery. EXPERT OPINION Aryl-urea bioisosteres of ω-3 PUFA epoxides that killed cancer cells targeted the mitochondrion by a novel dual mechanism: as protonophoric uncouplers and as inhibitors of electron transport complex III that activated ER-stress and disrupted mitochondrial integrity. In contrast, aryl-ureas that contain electron-donating substituents prevented cancer cell migration. Thus, aryl-ureas represent a novel class of agents with tunable anticancer properties. Stabilized analogues of other ω-3 PUFA-derived mediators could also be adapted into anticancer strategies. Indeed, a cocktail of agents that simultaneously promote cell killing, inhibit metastasis and angiogenesis, and that attenuate the pro-inflammatory microenvironment is a novel future anticancer strategy. Such regimen may enhance anticancer drug efficacy, minimize the development of anticancer drug resistance and enhance outcomes.
Collapse
Affiliation(s)
- Michael Murray
- Sydney Pharmacy School, Faculty of Medicine and Health, University of Sydney, NSW, Australia
- Woolcock Institute of Medical Research, Macquarie University, Macquarie Park, NSW, Australia
| |
Collapse
|
46
|
Kanefsky J, Basse M, Sokei J, di Martino O, Valin L, Jaspers Y, Martinez E, Huhn J, Di Marcantonio D, Magee JA, Goldman AR, Tang HY, Ferraro F, Kemp S, Wiest DL, Sykes SM. Disruption of polyunsaturated fatty acid biosynthesis drives STING-dependent acute myeloid leukemia cell maturation and death. J Biol Chem 2024; 300:107214. [PMID: 38522521 PMCID: PMC11061745 DOI: 10.1016/j.jbc.2024.107214] [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: 03/06/2023] [Revised: 02/20/2024] [Accepted: 03/13/2024] [Indexed: 03/26/2024] Open
Abstract
The role of polyunsaturated fatty acid (PUFA) biosynthesis in acute myeloid leukemia (AML) remains largely undefined. A comparative expression analysis of 35 genes encoding fatty acid biosynthesis enzymes showed that fatty acid desaturase 1 (FADS1) was highly expressed across multiple AML subtypes relative to healthy controls and that elevated FADS1 expression correlates with worse overall AML patient survival. Functionally, shRNA-mediated inhibition of FADS1 reduced AML cell growth in vitro and significantly delayed leukemia onset in an AML mouse model. AML cell lines depleted of FADS1 arrested in the G1/S-phase of the cell cycle, acquired characteristics of myeloid maturation and subsequently died. To understand the molecular consequences of FADS1 inhibition, a combination of mass spectrometry-based analysis of complex lipids and gene expression analysis (RNA-seq) was performed. FADS1 inhibition caused AML cells to exhibit significant lipidomic remodeling, including depletion of PUFAs from the phospholipids, phosphatidylserine, and phosphatidylethanolamine. These lipidomic alterations were accompanied by an increase induction of inflammatory and stimulator of interferon genes (STING)-mediated type-1 interferon signaling. Remarkably, genetic deletion of STING largely prevented the AML cell maturation and death phenotypes mediated by FADS1 inhibition. Highlighting the therapeutic implications of these findings, pharmacological blockade of PUFA biosynthesis reduced patient-derived AML cell numbers ex vivo but not that of healthy donor cells. Similarly, STING agonism attenuated patient-derived-AML survival; however, STING activation also reduced healthy granulocyte numbers. Collectively, these data unveil a previously unrecognized importance of PUFA biosynthesis in leukemogenesis and that imbalances in PUFA metabolism can drive STING-mediated AML maturation and death.
Collapse
Affiliation(s)
- Joice Kanefsky
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, USA
| | - Mary Basse
- Department of Medicine, School of Medicine, Washington University in Saint Louis, St Louis, Missouri, USA
| | - Judith Sokei
- Department of Medicine, School of Medicine, Washington University in Saint Louis, St Louis, Missouri, USA
| | - Orsola di Martino
- Department of Medicine, School of Medicine, Washington University in Saint Louis, St Louis, Missouri, USA
| | - Liana Valin
- Department of Medicine, School of Medicine, Washington University in Saint Louis, St Louis, Missouri, USA
| | - Yorrick Jaspers
- Amsterdam University Medical Centers, Amsterdam, North Holland, Netherlands
| | - Esteban Martinez
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, USA
| | - Jacklyn Huhn
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, USA
| | - Daniela Di Marcantonio
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, USA
| | - Jeffrey A Magee
- Department of Medicine, School of Medicine, Washington University in Saint Louis, St Louis, Missouri, USA
| | - Aaron R Goldman
- Proteomics & Metabolomics Facility, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Hsin-Yao Tang
- Proteomics & Metabolomics Facility, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Francesca Ferraro
- Department of Medicine, School of Medicine, Washington University in Saint Louis, St Louis, Missouri, USA
| | - Stephan Kemp
- Amsterdam University Medical Centers, Amsterdam, North Holland, Netherlands
| | - David L Wiest
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, USA.
| | - Stephen M Sykes
- Department of Medicine, School of Medicine, Washington University in Saint Louis, St Louis, Missouri, USA.
| |
Collapse
|
47
|
Xiang S, Jian Q, Chen W, Xu Q, Li J, Wang C, Wang R, Zhang D, Lin J, Zheng C. Pharmacodynamic components and mechanisms of ginger (Zingiber officinale) in the prevention and treatment of colorectal cancer. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117733. [PMID: 38218504 DOI: 10.1016/j.jep.2024.117733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/28/2023] [Accepted: 01/06/2024] [Indexed: 01/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginger is a "medicine-food homology" natural herb and has a longstanding medicinal background in treating intestinal diseases. Its remarkable bioactivities, including anti-inflammatory, antioxidant, immunoregulatory, flora regulatory, intestinal protective, and anticancer properties, make it a promising natural medicine for colorectal cancer (CRC) prevention and treatment. AIM OF THE REVIEW The purpose is to review the relevant literature on ginger and pharmacodynamic components for CRC prevention and treatment, summarize the possible mechanisms of ginger from clinical studies and animal and in vitro experiments, to provide theoretical support for the use of ginger preparations in the daily prevention and clinical treatment of CRC. MATERIALS AND METHODS Literatures about ginger and CRC were searched from electronic databases, such as PubMed, Web of Science, ScienceDirect, Google Scholar and China National Knowledge Infrastructure (CNKI). RESULTS This article summarizes the molecular mechanisms of ginger and its pharmacodynamic components in the prevention and treatment of CRC, including anti-inflammatory, antioxidant, immunoregulatory, flora regulatory, intestinal protective, inhibit CRC cell proliferation, induce CRC cell cycle blockage, promote CRC cell apoptosis, suppress CRC cell invasion and migration, enhance the anticancer effect of chemotherapeutic drugs. CONCLUSIONS Ginger has potential for daily prevention and clinical treatment of CRC.
Collapse
Affiliation(s)
- Sirui Xiang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Qin Jian
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Wu Chen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Qi Xu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jia Li
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Chuchu Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Rongrong Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Dingkun Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Junzhi Lin
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Chuan Zheng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
48
|
ZHAO J, ZHU Q, ZHANG Y, LI G, ZHANG Y, LI F, BIAN L. [Role of COX-2/PGE2/EP4 Axis-induced Macrophage Functional Activation
in NSCLC Development]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:245-256. [PMID: 38769827 PMCID: PMC11110263 DOI: 10.3779/j.issn.1009-3419.2024.101.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Indexed: 05/22/2024]
Abstract
BACKGROUND Tumor microenvironment (TME) is one of the important factors in tumorigenesis and progression, in which tumor-associated macrophages (TAMs) play an important role in non-small cell lung cancer (NSCLC) progression. However, the mechanism of TAMs in NSCLC progression remains unclear, so this study aimed to investigate the role of TAMs in NSCLC progression and to find potential therapeutic targets. METHODS Gene Expression Profiling Interactive Analysis (GEPIA) database was used to analyze the expression of prostaglandin E2 receptor 4 (EP4) mRNA in NSCLC and normal lung tissues; the protein expression levels of cyclooxygenase-2 (COX-2), EP4, cluster of differentiation 86 (CD86), CD163 and CD31 were detected by immunohistochemistry (IHC) in 120 NSCLC tissues and 24 paracancerous tissues specimens. The nude mouse lung adenocarcinoma cell A549 and macrophage RAW264.7 co-transplanted tumor model was established. And the samples were collected by gavage with EP4 inhibitor E7046, and then stained with hematoxylin-eosin (HE), IHC, and immunofluorescence (IF), and then detected by Western blot for the epithelial mesenchymal transformation (EMT) of the tumor tissues of the nude mice in each group. Western blot was used to detect the expressions of EMT related protiens in each group of nude mice; full-length transcriptome sequencing was used to screen the key genes causing liver metastasis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis was performed. RESULTS EP4 mRNA expression level in NSCLC tissues was generally lower than that in normal lung tissues (P<0.05); COX-2, EP4, CD163, CD31 proteins were differentially expressed in NSCLC tissues and adjacent tissues, and differences were observed in many clinicopathological parameters of NSCLC patients; RAW264.7 shortened the latency period of tumorigenesis of A549 and promoted the proliferation of tumors and liver metastasis of tumors, and E7046 could reduce tumor cell proliferation activity, tumor tissue vascular density and M2-type macrophage infiltration in nude mice; IF staining showed that macrophages were mainly distributed around the metastatic foci of tumors; Western blot results showed that compared with A549 alone transplantation group, the relative expression of E-cadherin protein in tumor tissues of mice in A549 and RAW264.7 co-transplantation group was significantly decreased, and the difference was statistically significant (P<0.05), while the relative expression of N-cadherin protein was up-regulated, but the difference was not statistically significant (P>0.05); the main pathways enriched in the differential genes of the full-length transcriptome were the PI3K-AKT and MAPK signaling pathways. CONCLUSIONS During NSCLC development, the COX-2/PGE2/EP4 axis may promote tumor progression by inducing macrophage functional activation, and EP4 may be a potential new target for tumor immunotherapy. This study provides new perspectives and ideas for in-depth exploration of the mechanisms of NSCLC development, as well as a theoretical basis for the development of new therapeutic strategies for NSCLC.
Collapse
|
49
|
Yadav P, Rana K, Nardini V, Khan A, Pani T, Kar A, Jain D, Chakraborty R, Singh R, Jha SK, Mehta D, Sharma H, Sharma RD, Deo SVS, Sengupta S, Patil VS, Faccioli LH, Dasgupta U, Bajaj A. Engineered nanomicelles inhibit the tumour progression via abrogating the prostaglandin-mediated immunosuppression. J Control Release 2024; 368:548-565. [PMID: 38462044 DOI: 10.1016/j.jconrel.2024.03.009] [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: 07/30/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024]
Abstract
Cancer treatment is challenged due to immunosuppressive inflammatory tumour microenvironment (TME) caused by infiltration of tumour-promoting and inhibition of tumour-inhibiting immune cells. Here, we report the engineering of chimeric nanomicelles (NMs) targeting the cell proliferation using docetaxel (DTX) and inflammation using dexamethasone (DEX) that alters the immunosuppressive TME. We show that a combination of phospholipid-DTX conjugate and PEGylated-lipid-DEX conjugate can self-assemble to form sub-100 nm chimeric NMs (DTX-DEX NMs). Anti-cancer activities against syngeneic and xenograft mouse models showed that the DTX-DEX NMs are more effective in tumour regression, enhance the survival of mice over other treatment modes, and alter the tumour stroma. DTX-DEX NMs cause a significant reduction in myeloid-derived suppressor cells, alter the polarization of macrophages, and enhance the accumulation of cytotoxic CD4+ and CD8+ T cells in tumour tissues, along with alterations in cytokine expression. We further demonstrated that these DTX-DEX NMs inhibit the synthesis of prostaglandins, especially PGE2, by targeting the cyclooxygenase 2 that is partly responsible for immunosuppressive TME. Therefore, this study presents, for the first time, the engineering of lithocholic acid-derived chimeric NMs that affect the prostaglandin pathway, alter the TME, and mitigate tumour progression with enhanced mice survival.
Collapse
Affiliation(s)
- Poonam Yadav
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Kajal Rana
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Viviani Nardini
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av do Café, s.n, Ribeirão Preto 14040-903, SP, Brazil
| | - Ali Khan
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon 122413, Haryana, India
| | - Trishna Pani
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon 122413, Haryana, India
| | - Animesh Kar
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Dolly Jain
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Ruchira Chakraborty
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Ragini Singh
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Somesh K Jha
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India
| | - Devashish Mehta
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon 122413, Haryana, India
| | - Harsh Sharma
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon 122413, Haryana, India
| | - Ravi Datta Sharma
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon 122413, Haryana, India
| | - S V S Deo
- Department of Surgical Oncology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sagar Sengupta
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India; National Institute of Biomedical Genomics, Post office- Netaji Subhas Sanatorium, Kalyani 741251, India
| | - Veena S Patil
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Lúcia Helena Faccioli
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av do Café, s.n, Ribeirão Preto 14040-903, SP, Brazil
| | - Ujjaini Dasgupta
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon 122413, Haryana, India
| | - Avinash Bajaj
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 3(rd) Milestone Faridabad-Gurgaon Expressway, NCR Biotech Cluster, Faridabad 121001, Haryana, India.
| |
Collapse
|
50
|
Kohno T, Kinoshita J, Oyama K, Saito H, Shimada M, Tsuji T, Yamamoto D, Moriyama H, Inaki N, Ohta T. Chemoprevention of esophageal adenocarcinoma in a rat surgical model by a cysteinyl leukotriene receptor‑1 antagonist. Oncol Lett 2024; 27:147. [PMID: 38385106 PMCID: PMC10879961 DOI: 10.3892/ol.2024.14280] [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: 10/20/2023] [Accepted: 01/25/2024] [Indexed: 02/23/2024] Open
Abstract
Reflux of gastroduodenal contents into the esophagus leads to the development of esophagitis and inflammation-associated pathologies, such as Barrett's esophagus (BE) and esophageal adenocarcinoma (EAC). The role of the lipoxygenase (LOX) pathway in carcinogenesis has been recently reported; however, its involvement in esophageal carcinogenesis remains unclear. To address this, the present study investigated the potential of pranlukast, a cysteinyl leukotriene receptor-1 antagonist, to suppress the progression of BE and EAC in a rat duodenogastroesophageal reflux (DGER) model. Male Wistar rats that underwent DGER were divided into two groups. One group was fed commercial chow (control group), and the other was fed experimental chow containing pranlukast (pranlukast group). The rats were sacrificed at 10, 20, 30 and 40 weeks after surgery, and their esophagi were examined. Expression levels of 5-LOX, CD68, IL-8, VEGF and Ki-67 were investigated using immunohistochemistry, and apoptosis was analyzed using the TUNEL method. In the pranlukast group, esophagitis was milder, and the incidence of BE and EAC was significantly lower (P<0.05) compared with that in the control group at 40 weeks after surgery. The number of cells positive for IL-8 and VEGF were significantly lower in the pranlukast group compared with the control group. Proliferative activity was also lower in the pranlukast group compared with the control group (P<0.05). Pranlukast treatment increased apoptosis (P<0.05). Overall, Pranlukast suppressed esophageal carcinogenesis in a rat DGER model, decreasing inflammatory cytokines such as IL-8 and VEGF.
Collapse
Affiliation(s)
- Tatsuhiko Kohno
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Jun Kinoshita
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Katsunobu Oyama
- Department of Surgery, Public Central Hospital of Matto Ishikawa, Hakusan, Ishikawa 924-0865, Japan
| | - Hiroto Saito
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Mari Shimada
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Toshikatsu Tsuji
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Daisuke Yamamoto
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Hideki Moriyama
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Noriyuki Inaki
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Tetsuo Ohta
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| |
Collapse
|