1
|
Gapińska N, Wlaź P, Wyska E, Świerczek A, Kamiński K, Jakubiec M, Abram M, Ciepiela K, Latacz G, Słowik T, Krokowski D, Jarosz Ł, Ciszewski A, Socała K. Effect of SSR504734, a Selective Glycine Transporter Type 1 Inhibitor, on Seizure Thresholds, Neurotransmitter Levels, and Inflammatory Markers in Mice. ACS Chem Neurosci 2025; 16:1210-1226. [PMID: 40012256 PMCID: PMC11926788 DOI: 10.1021/acschemneuro.5c00039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
Studies have revealed that inhibition of glycine transporter type 1 (GlyT1) may provide a balanced regulation between excitation and inhibition in some brain structures and, thereby, modulate seizure activity. Data on the role of GlyT1 in epilepsy are, however, very limited. Here, we examined the effect of SSR504734, a highly selective and reversible GlyT1 inhibitor, on three acute seizure tests in mice. We also evaluated its impact on neurotransmitter levels in the relevant brain structures following seizures, possible adverse effects, and changes in the levels of inflammatory mediators in the serum and liver. In addition, in vivo pharmacokinetic profile and in vitro ADME-Tox properties of SSR504734 were investigated. The results show that SSR504734 significantly increased the threshold for tonic hindlimb extension in the MEST test after acute and repeated treatment but had no influence on seizure thresholds in the 6 Hz and i.v. PTZ seizure tests. SSR504734 did not affect the levels of glutamate, GABA, glycine, or adenosine in brain structures of mice with MES-induced seizures. However, after acute treatment, the concentration of glutamate and adenosine in the brainstem of control animals (i.e., without seizures) decreased. Moreover, SSR504734 increased the levels of inflammatory markers (TNF-α, Il-1β, IL-6, IL-10, and TLR4) in serum. In vivo pharmacokinetic profiling and in vitro ADME-Tox data confirmed suitable drug-like properties of SSR504734, including its notable penetration into brain tissue. However, possible hepatotoxicity at higher doses should be taken into account. Further studies should be considered to better characterize the SSR504734-mediated effects as well as to validate GlyT1 as a potential new molecular target in epilepsy treatment.
Collapse
Affiliation(s)
- Nikola Gapińska
- Department
of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
- Doctoral
School of Quantitative and Natural Sciences, Maria Curie-Skłodowska University, Weteranów 18, 20-038 Lublin, Poland
| | - Piotr Wlaź
- Department
of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Elżbieta Wyska
- Department
of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Artur Świerczek
- Department
of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Krzysztof Kamiński
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Marcin Jakubiec
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Michał Abram
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Katarzyna Ciepiela
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
- Selvita
S.A., Bobrzyńskiego
14, 30-348 Cracow, Poland
| | - Gniewomir Latacz
- Department
of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Tymoteusz Słowik
- Experimental
Medicine Center, Medical University, Jaczewskiego 8, 20-090 Lublin, Poland
| | - Dawid Krokowski
- Department
of Molecular Biology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Łukasz Jarosz
- Department
of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary
Medicine, University of Life Sciences in
Lublin, Głęboka 30, 20-612 Lublin, Poland
| | - Artur Ciszewski
- Department
of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary
Medicine, University of Life Sciences in
Lublin, Głęboka 30, 20-612 Lublin, Poland
| | - Katarzyna Socała
- Department
of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| |
Collapse
|
2
|
Johnson E, Albakri JS, Allemailem KS, Sultan A, Alwanian WM, Alrumaihi F, Almansour NM, Aldakheel FM, Khalil FMA, Abduallah AM, Smith O. Mitochondrial dysfunction and calcium homeostasis in heart failure: Exploring the interplay between oxidative stress and cardiac remodeling for future therapeutic innovations. Curr Probl Cardiol 2025; 50:102968. [PMID: 39653095 DOI: 10.1016/j.cpcardiol.2024.102968] [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: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
Heart failure (HF) is a multifaceted clinical syndrome characterized by the heart's inability to pump sufficient blood to meet the body's metabolic demands. It arises from various etiologies, including myocardial injury, hypertension, and valvular heart disease. A critical aspect of HF pathophysiology involves mitochondrial dysfunction, particularly concerning calcium (Ca2+) homeostasis and oxidative stress. This review highlights the pivotal role of excess mitochondrial Ca2+ in exacerbating oxidative stress, contributing significantly to HF progression. Novel insights are provided regarding the mechanisms by which mitochondrial Ca2+ overload leads to increased production of reactive oxygen species (ROS) and impaired cellular function. Despite this understanding, key gaps in research remain, particularly in elucidating the complex interplay between mitochondrial dynamics and oxidative stress across different HF phenotypes. Furthermore, therapeutic strategies targeting mitochondrial dysfunction are still in their infancy, with limited applications in clinical practice. By summarizing recent findings and identifying these critical research gaps, this review aims to pave the way for innovative therapeutic approaches that improve the management of heart failure, ultimately enhancing patient outcomes through targeted interventions.
Collapse
Affiliation(s)
- Emily Johnson
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | | | - Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Abdulaziz Sultan
- Family Medicine Senior Registrar, Ministry of Health, Saudi Arabia
| | - Wanian M Alwanian
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Nahlah Makki Almansour
- Department of Biology, College of Science, University of Hafr Al Batin, Hafr Al Batin 31991, Saudi Arabia
| | - Fahad M Aldakheel
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Fatma Mohamed Ameen Khalil
- King Khalid University, Applied College, Unit of health specialties, basic sciences and their applications, Mohayil Asir Abha, 61421, Saudi Arabia
| | - Alduwish Manal Abduallah
- Department of Biology, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Alkarj 11942, Saudi Arabia
| | - Oliver Smith
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
3
|
Yang H, Atak D, Yuan M, Li M, Altay O, Demirtas E, Peltek IB, Ulukan B, Yigit B, Sipahioglu T, Álvez MB, Meng L, Yüksel B, Turkez H, Kirimlioglu H, Saka B, Yurdaydin C, Akyildiz M, Dayangac M, Uhlen M, Boren J, Zhang C, Mardinoglu A, Zeybel M. Integrative proteo-transcriptomic characterization of advanced fibrosis in chronic liver disease across etiologies. Cell Rep Med 2025; 6:101935. [PMID: 39889710 PMCID: PMC11866494 DOI: 10.1016/j.xcrm.2025.101935] [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: 05/29/2024] [Revised: 09/20/2024] [Accepted: 01/08/2025] [Indexed: 02/03/2025]
Abstract
Chronic hepatic injury and inflammation from various causes can lead to fibrosis and cirrhosis, potentially predisposing to hepatocellular carcinoma. The molecular mechanisms underlying fibrosis and its progression remain incompletely understood. Using a proteo-transcriptomics approach, we analyze liver and plasma samples from 330 individuals, including 40 healthy individuals and 290 patients with histologically characterized fibrosis due to chronic viral infection, alcohol consumption, or metabolic dysfunction-associated steatotic liver disease. Our findings reveal dysregulated pathways related to extracellular matrix, immune response, inflammation, and metabolism in advanced fibrosis. We also identify 132 circulating proteins associated with advanced fibrosis, with neurofascin and growth differentiation factor 15 demonstrating superior predictive performance for advanced fibrosis(area under the receiver operating characteristic curve [AUROC] 0.89 [95% confidence interval (CI) 0.81-0.97]) compared to the fibrosis-4 model (AUROC 0.85 [95% CI 0.78-0.93]). These findings provide insights into fibrosis pathogenesis and highlight the potential for more accurate non-invasive diagnosis.
Collapse
Affiliation(s)
- Hong Yang
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Dila Atak
- Department of Gastroenterology and Hepatology, School of Medicine, Koç University, İstanbul 34010, Turkiye
| | - Meng Yuan
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Mengzhen Li
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Ozlem Altay
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Elif Demirtas
- School of Medicine, Koç University, Istanbul 34450, Turkiye
| | | | - Burge Ulukan
- Department of Gastroenterology and Hepatology, School of Medicine, Koç University, İstanbul 34010, Turkiye
| | - Buket Yigit
- Department of Gastroenterology and Hepatology, School of Medicine, Koç University, İstanbul 34010, Turkiye
| | - Tarik Sipahioglu
- Department of Gastroenterology and Hepatology, School of Medicine, Koç University, İstanbul 34010, Turkiye
| | - María Bueno Álvez
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Lingqi Meng
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | | | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkiye
| | - Hale Kirimlioglu
- Department of Pathology, School of Medicine, Acibadem Mehmet Ali Aydinlar University Istanbul 34752, Turkiye
| | - Burcu Saka
- Department of Pathology, School of Medicine, Koç University, Istanbul 34010, Turkiye
| | - Cihan Yurdaydin
- Department of Gastroenterology and Hepatology, School of Medicine, Koç University, İstanbul 34010, Turkiye
| | - Murat Akyildiz
- Department of Gastroenterology and Hepatology, School of Medicine, Koç University, İstanbul 34010, Turkiye
| | - Murat Dayangac
- Department of General Surgery, International School of Medicine, Medipol University, Istanbul 34010, Turkiye
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Jan Boren
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, UK.
| | - Mujdat Zeybel
- Department of Gastroenterology and Hepatology, School of Medicine, Koç University, İstanbul 34010, Turkiye; Clinical Trials Unit, Koç University Hospital, Istanbul 34010, Turkiye.
| |
Collapse
|
4
|
Jiang Y, Tao Q, Qiao X, Yang Y, Peng C, Han M, Dong K, Zhang W, Xu M, Wang D, Zhu W, Li X. Targeting amino acid metabolism to inhibit gastric cancer progression and promote anti-tumor immunity: a review. Front Immunol 2025; 16:1508730. [PMID: 40018041 PMCID: PMC11864927 DOI: 10.3389/fimmu.2025.1508730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/24/2025] [Indexed: 03/01/2025] Open
Abstract
The incidence of gastric cancer remains high and poses a serious threat to human health. Recent comprehensive investigations into amino acid metabolism and immune system components within the tumor microenvironment have elucidated the functional interactions between tumor cells, immune cells, and amino acid metabolism. This study reviews the characteristics of amino acid metabolism in gastric cancer, with a particular focus on the metabolism of methionine, cysteine, glutamic acid, serine, taurine, and other amino acids. It discusses the relationship between these metabolic processes, tumor development, and the body's anti-tumor immunity, and analyzes the importance of targeting amino acid metabolism in gastric cancer for chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Yuchun Jiang
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Qing Tao
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xuehan Qiao
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yufei Yang
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chen Peng
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Miao Han
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Kebin Dong
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wei Zhang
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, China
| | - Min Xu
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, China
| | - Deqiang Wang
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, China
| | - Wen Zhu
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaoqin Li
- Department of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
5
|
Shin KWD, Atalay MV, Cetin-Atalay R, O'Leary EM, Glass ME, Szafran JCH, Woods PS, Meliton AY, Shamaa OR, Tian Y, Mutlu GM, Hamanaka RB. mTOR signaling regulates multiple metabolic pathways in human lung fibroblasts after TGF-β and in pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2025; 328:L215-L228. [PMID: 39745695 DOI: 10.1152/ajplung.00189.2024] [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: 06/20/2024] [Revised: 11/25/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025] Open
Abstract
Idiopathic pulmonary fibrosis is a fatal disease characterized by the transforming growth factor (TGF-β)-dependent activation of lung fibroblasts, leading to excessive deposition of collagen proteins and progressive replacement of healthy lungs with scar tissue. We and others have shown that TGF-β-mediated activation of the mechanistic target of rapamycin complex 1 (mTORC1) and downstream upregulation of activating transcription factor 4 (ATF4) promotes metabolic reprogramming in lung fibroblasts characterized by upregulation of the de novo synthesis of glycine, the most abundant amino acid found in collagen protein. Whether mTOR and ATF4 regulate other metabolic pathways in lung fibroblasts has not been explored. Here, we used RNA sequencing to determine how both ATF4 and mTOR regulate gene expression in human lung fibroblasts following TGF-β. We found that ATF4 primarily regulates enzymes and transporters involved in amino acid homeostasis as well as aminoacyl-tRNA synthetases. mTOR inhibition resulted not only in the loss of ATF4 target gene expression but also in the reduced expression of glycolytic enzymes and mitochondrial electron transport chain subunits. Analysis of TGF-β-induced changes in cellular metabolite levels confirmed that ATF4 regulates amino acid homeostasis in lung fibroblasts, whereas mTOR also regulates glycolytic and TCA cycle metabolites. We further analyzed publicly available single-cell RNA-seq datasets and found increased expression of ATF4 and mTOR-regulated genes in pathologic fibroblast populations from the lungs of patients with IPF. Our results provide insight into the mechanisms of metabolic reprogramming in lung fibroblasts and highlight novel ATF4 and mTOR-dependent pathways that may be targeted to inhibit fibrotic processes.NEW & NOTEWORTHY Here, we used transcriptomic and metabolomic approaches to develop a more complete understanding of the role that mTOR, and its downstream effector ATF4, play in promoting metabolic reprogramming in lung fibroblasts. We identify novel metabolic pathways that may promote pathologic phenotypes, and we provide evidence from single-cell RNA-seq datasets that similar metabolic reprogramming occurs in patient lungs.
Collapse
Affiliation(s)
- Kun Woo D Shin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - M Volkan Atalay
- Department of Information Systems and Supply Chain Management, Loyola University Chicago, Chicago, Illinois, United States
| | - Rengul Cetin-Atalay
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Erin M O'Leary
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Mariel E Glass
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Jennifer C Houpy Szafran
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Parker S Woods
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Angelo Y Meliton
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Obada R Shamaa
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Yufeng Tian
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Gökhan M Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Robert B Hamanaka
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| |
Collapse
|
6
|
Wu Y, Millender J, Padgett B, Marx M, Madnick S, Puterbaugh R, Angelo KS, Hopkins CM, Morgan JR. An in vitro model to measure the strength and stiffness of the extracellular matrix synthesized de novo by human fibroblasts. IN VITRO MODELS 2025; 4:59-69. [PMID: 40160211 PMCID: PMC11950444 DOI: 10.1007/s44164-025-00081-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 04/02/2025]
Abstract
Purpose Alterations to the strength and stiffness of the human extracellular matrix (ECM) are the underlying pathology manifest in a wide range of diseases. These include inherited conditions, such as Ehlers Danlos syndrome, as well as acquired diseases such as fibrosis, which remains a major unmet medical need. To evaluate promising therapies, new models are needed that can measure the strength and stiffness of the human ECM. Methods Cultured human fibroblasts were seeded into circular troughs of agarose that had been molded into a 24 well plate and equilibrated with cell culture medium. The cells settled by gravity, aggregated and formed 3D ring-shaped tissues 5 mm in diameter without the aid of added exogenous scaffold material. The ECM proteins synthesized de novo by the rings were characterized by immuno-staining. The response of the rings to drug and growth factor treatments were assessed by measuring changes to the dimensions of the rings and by measuring levels of collagen. A tensile test was used to quantify drug and growth factor induced changes to the strength and stiffness of the rings. Results Ring-shaped tissues readily formed in the molds and synthesized de novo a circumferentially aligned collagen-rich fibrous ECM network positive for collagen type I, collagen type III and fibronectin. Low dose treatment with incyclinide, an inhibitor of matrix metalloproteinases (MMPs), increased strength and stiffness, whereas as a high dose decreased tensile properties, likely due to a toxic effect. Treatment with TGF-β1, a well-known driver of fibrosis, increased levels of collagen and tensile properties and mimicked the fibrotic environment in vitro. Treatment with PAT-1251, an inhibitor of the collagen crosslinking enzyme lysyl oxidase-like protein 2 (LOXL2), had no effect on levels of collagen but significantly reduced the strength and stiffness of the ring even when elevated by treatment with TGF-β1. Conclusion Human fibroblasts will self-assemble a 3D ring-shaped tissue and synthesize a fibrous network of ECM proteins whose tensile properties can be measured. The fibrotic environment can be mimicked by addition of TGF-β1, which increases levels of collagen as well as the strength and stiffness of the rings. Treatment with two drugs, incyclinide and PAT 1251 that were developed as potential treatments for diseases of the ECM, altered the strength and stiffness of the rings, thereby demonstrating the utility of the model for testing new therapies that target the biomechanics of the ECM. Supplementary Information The online version contains supplementary material available at 10.1007/s44164-025-00081-y.
Collapse
Affiliation(s)
- Yanying Wu
- Department of Pathology and Laboratory Medicine, Brown University, 171 Meeting St, Providence, G-B 393 USA
- Center for Alternatives to Animals in Testing, Brown University, Providence, RI USA
| | - Jayla Millender
- Department of Pathology and Laboratory Medicine, Brown University, 171 Meeting St, Providence, G-B 393 USA
- Center for Alternatives to Animals in Testing, Brown University, Providence, RI USA
| | - Becka Padgett
- Department of Pathology and Laboratory Medicine, Brown University, 171 Meeting St, Providence, G-B 393 USA
- Center for Alternatives to Animals in Testing, Brown University, Providence, RI USA
| | - Madeleine Marx
- Department of Pathology and Laboratory Medicine, Brown University, 171 Meeting St, Providence, G-B 393 USA
- Center for Alternatives to Animals in Testing, Brown University, Providence, RI USA
| | - Samantha Madnick
- Department of Pathology and Laboratory Medicine, Brown University, 171 Meeting St, Providence, G-B 393 USA
- Center for Alternatives to Animals in Testing, Brown University, Providence, RI USA
| | - Ryan Puterbaugh
- Department of Pathology and Laboratory Medicine, Brown University, 171 Meeting St, Providence, G-B 393 USA
- Center for Alternatives to Animals in Testing, Brown University, Providence, RI USA
| | - Katerina St. Angelo
- Department of Pathology and Laboratory Medicine, Brown University, 171 Meeting St, Providence, G-B 393 USA
- Center for Alternatives to Animals in Testing, Brown University, Providence, RI USA
| | - Caitlin M. Hopkins
- Department of Pathology and Laboratory Medicine, Brown University, 171 Meeting St, Providence, G-B 393 USA
- Center for Alternatives to Animals in Testing, Brown University, Providence, RI USA
| | - Jeffrey R. Morgan
- Department of Pathology and Laboratory Medicine, Brown University, 171 Meeting St, Providence, G-B 393 USA
- Center for Alternatives to Animals in Testing, Brown University, Providence, RI USA
| |
Collapse
|
7
|
Sun W, Zhou S, Peng L, Wang W, Liu Y, Wang T, Cheng D, Li Z, Xiong H, Jia X, Lian W, Jiao J, Ni C. Fatty Acid Oxidation-Glycolysis Metabolic Transition Affects ECM Homeostasis in Silica-Induced Pulmonary Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407134. [PMID: 39721015 PMCID: PMC11831484 DOI: 10.1002/advs.202407134] [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: 06/26/2024] [Revised: 11/22/2024] [Indexed: 12/28/2024]
Abstract
Silicosis is a fatal occupational pulmonary disease that is characterized by irreversible replacement of lung parenchyma by aberrant Exracellular matrix (ECM). Metabolic reprogramming is a crucial mechanism for fibrosis. However, how the metabolic rewiring shifts the ECM homeostasis toward overaccumulation remains unclear. Herein, a phenotype with reduction in fatty acid oxidation (FAO) but enhanced glycolysis in myofibroblasts is shown. Perturbation of the glycolytic and FAO pathways, respectively, reveals distinct roles in the metabolic distribution of ECM deposition and degradation. Suppressed glycolysis leads to a decrease in insoluble ECM, primarily due to the inhibition of ECM-modifying enzyme activity and a decrease in glycine synthesis. Notably, promoted FAO facilitates the intracellular degradation pathway of ECM. In addition, the findings revealed that hypoxia-inducible factor-1 alpha (HIF-1α) serves as a crucial metabolic regulator in the transition from FAO to glycolysis, thereby playing a significant role in ECM deposition in silica-induced pulmonary fibrosis. Further, the promotion of FAO, inhibition of glycolysis and HIF-1α reduce ECM production and promote ECM degradation, ultimately impeding the progression of fibrosis and providing therapeutic relief for established pulmonary fibrosis in vivo. These findings unveil the metabolic rewire underpinning the deposition of ECM in silica-induced lung fibrosis and identify novel targets for promoting regression of pulmonary fibrosis.
Collapse
Affiliation(s)
- Wenqing Sun
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical UniversityWuxi Center for Disease Control and PreventionWuxi Medical CenterNanjing medical universityNanjing211166China
| | - Siyun Zhou
- Department of Occupational Medical and Environmental HealthKey Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjing211166China
| | - Lan Peng
- Department of Occupational Medical and Environmental HealthKey Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjing211166China
| | - Wei Wang
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical UniversityWuxi Center for Disease Control and PreventionWuxi Medical CenterNanjing medical universityNanjing211166China
| | - Yi Liu
- Department of Occupational Medical and Environmental HealthKey Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjing211166China
| | - Ting Wang
- Department of Occupational Medical and Environmental HealthKey Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjing211166China
- Department of PathologyNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing210000China
| | - Demin Cheng
- Department of Occupational Medical and Environmental HealthKey Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjing211166China
| | - Ziwei Li
- Department of Occupational Medical and Environmental HealthKey Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjing211166China
| | - Haojie Xiong
- Department of Occupational Medical and Environmental HealthKey Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjing211166China
| | - Xinying Jia
- Department of Occupational Medical and Environmental HealthKey Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjing211166China
| | - Wenxiu Lian
- Department of Occupational Medical and Environmental HealthKey Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjing211166China
| | - Jiandong Jiao
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical UniversityWuxi Center for Disease Control and PreventionWuxi Medical CenterNanjing medical universityNanjing211166China
| | - Chunhui Ni
- Department of Occupational Medical and Environmental HealthKey Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjing211166China
- Department of Public HealthKangda College of Nanjing Medical UniversityLianyungang320700China
| |
Collapse
|
8
|
Hansman DS, Du J, Casson RJ, Peet DJ. Eye on the horizon: The metabolic landscape of the RPE in aging and disease. Prog Retin Eye Res 2025; 104:101306. [PMID: 39433211 PMCID: PMC11833275 DOI: 10.1016/j.preteyeres.2024.101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
To meet the prodigious bioenergetic demands of the photoreceptors, glucose and other nutrients must traverse the retinal pigment epithelium (RPE), a polarised monolayer of cells that lie at the interface between the outer retina and the choroid, the principal vascular layer of the eye. Recent investigations have revealed a metabolic ecosystem in the outer retina where the photoreceptors and RPE engage in a complex exchange of sugars, amino acids, and other metabolites. Perturbation of this delicate metabolic balance has been identified in the aging retina, as well as in age-related macular degeneration (AMD), the leading cause of blindness in the Western world. Also common in the aging and diseased retina are elevated levels of cytokines, oxidative stress, advanced glycation end-products, increased growth factor signalling, and biomechanical stress - all of which have been associated with metabolic dysregulation in non-retinal cell types and tissues. Herein, we outline the role of these factors in retinal homeostasis, aging, and disease. We discuss their effects on glucose, mitochondrial, lipid, and amino acid metabolism in tissues and cell types outside the retina, highlighting the signalling pathways through which they induce these changes. Lastly, we discuss promising avenues for future research investigating the roles of these pathological conditions on retinal metabolism, potentially offering novel therapeutic approaches to combat age-related retinal disease.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
9
|
Guillard J, Schwörer S. Metabolic control of collagen synthesis. Matrix Biol 2024; 133:43-56. [PMID: 39084474 PMCID: PMC11402592 DOI: 10.1016/j.matbio.2024.07.003] [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: 05/26/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
The extracellular matrix (ECM) is present in all tissues and crucial in maintaining normal tissue homeostasis and function. Defects in ECM synthesis and remodeling can lead to various diseases, while overproduction of ECM components can cause severe conditions like organ fibrosis and influence cancer progression and therapy resistance. Collagens are the most abundant core ECM proteins in physiological and pathological conditions and are predominantly synthesized by fibroblasts. Previous efforts to target aberrant collagen synthesis in fibroblasts by inhibiting pro-fibrotic signaling cascades have been ineffective. More recently, metabolic rewiring downstream of pro-fibrotic signaling has emerged as a critical regulator of collagen synthesis in fibroblasts. Here, we propose that targeting the metabolic pathways involved in ECM biomass generation provides a novel avenue for treating conditions characterized by excessive collagen accumulation. This review summarizes the unique metabolic challenges collagen synthesis imposes on fibroblasts and discusses how underlying metabolic networks could be exploited to create therapeutic opportunities in cancer and fibrotic disease. Finally, we provide a perspective on open questions in the field and how conceptual and technical advances will help address them to unlock novel metabolic vulnerabilities of collagen synthesis in fibroblasts and beyond.
Collapse
Affiliation(s)
- Julien Guillard
- Section of Hematology/Oncology, Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, 60637, USA
| | - Simon Schwörer
- Section of Hematology/Oncology, Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, 60637, USA; Committee on Cancer Biology, Committee on Molecular Metabolism and Nutrition, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
10
|
Chen H, Xu X, Li J, Xue Y, Li X, Zhang K, Jiang H, Liu X, Li M. Decoding tumor-fibrosis interplay: mechanisms, impact on progression, and innovative therapeutic strategies. Front Pharmacol 2024; 15:1491400. [PMID: 39534084 PMCID: PMC11555290 DOI: 10.3389/fphar.2024.1491400] [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: 09/04/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Malignant tumors are a category of diseases that possess invasive and metastatic capabilities, with global incidence and mortality rates remaining high. In recent years, the pivotal role of fibrosis in tumor progression, drug resistance, and immune evasion has increasingly been acknowledged. Fibrosis enhances the proliferation, migration, and invasion of tumor cells by modifying the composition and structure of the extracellular matrix, thereby offering protection for immune evasion by tumor cells. The activation of cancer-associated fibroblasts (CAFs) plays a significant role in this process, as they further exacerbate the malignant traits of tumors by secreting a variety of cytokines and growth factors. Anti-fibrotic tumor treatment strategies, including the use of anti-fibrotic drugs and inhibition of fibrosis-related signaling pathways such as Transforming Growth Factor-β (TGF-β), have demonstrated potential in delaying tumor progression and improving the effectiveness of chemotherapy, targeted therapy, and immunotherapy. In the future, by developing novel drugs that target the fibrotic microenvironment, new therapeutic options may be available for patients with various refractory tumors.
Collapse
Affiliation(s)
- Huiguang Chen
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xuexin Xu
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Jingxian Li
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Yu Xue
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xin Li
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Kaiyu Zhang
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Haihui Jiang
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaoliu Liu
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
- Department of Anatomy, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Mingzhe Li
- Department of Anatomy, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Zhou Z, Xie Y, Wei Q, Zhang X, Xu Z. Revisiting the role of MicroRNAs in the pathogenesis of idiopathic pulmonary fibrosis. Front Cell Dev Biol 2024; 12:1470875. [PMID: 39479511 PMCID: PMC11521927 DOI: 10.3389/fcell.2024.1470875] [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: 07/26/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a prevalent chronic pulmonary fibrosis disease characterized by alveolar epithelial cell damage, fibroblast proliferation and activation, excessive extracellular matrix deposition, and abnormal epithelial-mesenchymal transition (EMT), resulting in tissue remodeling and irreversible structural distortion. The mortality rate of IPF is very high, with a median survival time of 2-3 years after diagnosis. The exact cause of IPF remains unknown, but increasing evidence supports the central role of epigenetic changes, particularly microRNA (miRNA), in IPF. Approximately 10% of miRNAs in IPF lung tissue exhibit differential expression compared to normal lung tissue. Diverse miRNA phenotypes exert either a pro-fibrotic or anti-fibrotic influence on the progression of IPF. In the context of IPF, epigenetic factors such as DNA methylation and long non-coding RNAs (lncRNAs) regulate differentially expressed miRNAs, which in turn modulate various signaling pathways implicated in this process, including transforming growth factor-β1 (TGF-β1)/Smad, mitogen-activated protein kinase (MAPK), and phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT) pathways. Therefore, this review presents the epidemiology of IPF, discusses the multifaceted regulatory roles of miRNAs in IPF, and explores the impact of miRNAs on IPF through various pathways, particularly the TGF-β1/Smad pathway and its constituent structures. Consequently, we investigate the potential for targeting miRNAs as a treatment for IPF, thereby contributing to advancements in IPF research.
Collapse
Affiliation(s)
| | | | | | | | - Zhihao Xu
- The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| |
Collapse
|
12
|
Yang Y, Wang W, Zeng Q, Wang N, Li W, Chen B, Guan Q, Li C, Li W. Fabricating oxygen self-supplying 3D printed bioactive hydrogel scaffold for augmented vascularized bone regeneration. Bioact Mater 2024; 40:227-243. [PMID: 38973993 PMCID: PMC11226730 DOI: 10.1016/j.bioactmat.2024.06.016] [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: 03/12/2024] [Revised: 05/26/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024] Open
Abstract
Limited cells and factors, inadequate mechanical properties, and necrosis of defects center have hindered the wide clinical application of bone-tissue engineering scaffolds. Herein, we construct a self-oxygenated 3D printed bioactive hydrogel scaffold by integrating oxygen-generating nanoparticles and hybrid double network hydrogel structure. The hydrogel scaffold possesses the characteristics of extracellular matrix; Meanwhile, the fabricated hybrid double network structure by polyacrylamide and CaCl2-crosslinked sodium carboxymethylcellulose endows the hydrogel favorable compressive strength and 3D printability. Furthermore, the O2 generated by CaO2 nanoparticles encapsulated in ZIF-8 releases steadily and sustainably because of the well-developed microporous structure of ZIF-8, which can significantly promote cell viability and proliferation in vitro, as well as angiogenesis and osteogenic differentiation with the assistance of Zn2+. More significantly, the synergy of O2 and 3D printed pore structure can prevent necrosis of defects center and facilitate cell infiltration by providing cells the nutrients and space they need, which can further induce vascular network ingrowth and accelerate bone regeneration in all areas of the defect in vivo. Overall, this work provides a new avenue for preparing cell/factor-free bone-tissue engineered scaffolds that possess great potential for tissue regeneration and clinical alternative.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Wanmeng Wang
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, School of Stomatology, Tianjin Medical University, Tianjin, 300071, PR China
| | - Qianrui Zeng
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Ning Wang
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, School of Stomatology, Tianjin Medical University, Tianjin, 300071, PR China
| | - Wenbo Li
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Bo Chen
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, School of Stomatology, Tianjin Medical University, Tianjin, 300071, PR China
| | - Qingxin Guan
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Changyi Li
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, School of Stomatology, Tianjin Medical University, Tianjin, 300071, PR China
| | - Wei Li
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, PR China
| |
Collapse
|
13
|
Ji YW, Wen XY, Tang HP, Jin ZS, Su WT, Zhou L, Xia ZY, Xia ZY, Lei SQ. DJ-1: Potential target for treatment of myocardial ischemia-reperfusion injury. Biomed Pharmacother 2024; 179:117383. [PMID: 39232383 DOI: 10.1016/j.biopha.2024.117383] [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: 06/21/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Ischemic heart disease (IHD) is a significant global health concern, resulting in high rates of mortality and disability among patients. Although coronary blood flow reperfusion is a key treatment for IHD, it often leads to acute myocardial ischemia-reperfusion injury (IRI). Current intervention strategies have limitations in providing adequate protection for the ischemic myocardium. DJ-1, originally known as a Parkinson's disease related protein, is a highly conserved cytoprotective protein. It is involved in enhancing mitochondrial function, scavenging reactive oxygen species (ROS), regulating autophagy, inhibiting apoptosis, modulating anaerobic metabolism, and exerting anti-inflammatory effects. DJ-1 is also required for protective strategies, such as ischemic preconditioning, ischemic postconditioning, remote ischemic preconditioning and pharmacological conditioning. Therefore, DJ-1 emerges as a potential target for the treatment of myocardial IRI. Our comprehensive review delves into its protective mechanisms in myocardial IRI and the structural foundations underlying its functions.
Collapse
Affiliation(s)
- Yan-Wei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin-Yu Wen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - He-Peng Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhen-Shuai Jin
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wa-Ting Su
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lu Zhou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zheng-Yuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shao-Qing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
14
|
Pan M, Li H, Shi X. A New Target for Hepatic Fibrosis Prevention and Treatment: The Warburg Effect. FRONT BIOSCI-LANDMRK 2024; 29:321. [PMID: 39344326 DOI: 10.31083/j.fbl2909321] [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: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 10/01/2024]
Abstract
Hepatic fibrosis is a major public health problem that endangers human wellbeing. In recent years, a number of studies have revealed the important impact of metabolic reprogramming on the occurrence and development of hepatic fibrosis. Among them, the Warburg effect, as an intracellular glucose metabolism reprogramming, can promote the occurrence and development of hepatic fibrosis by promoting the activation of hepatic stellate cells (HSCs) and inducing the polarization of liver macrophages (KC). Understanding the Warburg effect and its important role in the progression of hepatic fibrosis will assist in developing new strategies for the prevention and treatment of hepatic fibrosis. This review focuses on the Warburg effect and the specific mechanism by which it affects the progression of hepatic fibrosis by regulating HSCs activation and KC polarization. In addition, we also summarize and discuss the related experimental drugs and their mechanisms that inhibit the Warburg effect by targeting key proteins of glycolysis in order to improve hepatic fibrosis in the hope of providing more effective strategies for the clinical treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Meng Pan
- College of Basic Medical Sciences, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| | - Huanyu Li
- Second Clinical Medical College, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| | - Xiaoyan Shi
- College of Basic Medical Sciences, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| |
Collapse
|
15
|
Liu C, Wei W, Huang Y, Fu P, Zhang L, Zhao Y. Metabolic reprogramming in septic acute kidney injury: pathogenesis and therapeutic implications. Metabolism 2024; 158:155974. [PMID: 38996912 DOI: 10.1016/j.metabol.2024.155974] [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: 04/10/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Acute kidney injury (AKI) is a frequent and severe complication of sepsis and is characterized by significant mortality and morbidity. However, the pathogenesis of septic acute kidney injury (S-AKI) remains elusive. Metabolic reprogramming, which was originally referred to as the Warburg effect in cancer, is strongly related to S-AKI. At the onset of sepsis, both inflammatory cells and renal parenchymal cells, such as macrophages, neutrophils and renal tubular epithelial cells, undergo metabolic shifts toward aerobic glycolysis to amplify proinflammatory responses and fortify cellular resilience to septic stimuli. As the disease progresses, these cells revert to oxidative phosphorylation, thus promoting anti-inflammatory reactions and enhancing functional restoration. Alterations in mitochondrial dynamics and metabolic reprogramming are central to the energetic changes that occur during S-AKI. In this review, we summarize the current understanding of the pathogenesis of metabolic reprogramming in S-AKI, with a focus on each cell type involved. By identifying relevant key regulatory factors, we also explored potential metabolic reprogramming-related therapeutic targets for the management of S-AKI.
Collapse
Affiliation(s)
- Caihong Liu
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Wei Wei
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yongxiu Huang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ping Fu
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ling Zhang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yuliang Zhao
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu 610041, China.
| |
Collapse
|
16
|
Alatawi S, Alzamzami W. New insights into PSAT1 as a therapeutic target for myelodysplastic syndrome (MDS). PLoS One 2024; 19:e0309456. [PMID: 39186541 PMCID: PMC11346733 DOI: 10.1371/journal.pone.0309456] [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: 05/23/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
The metabolomic landscape in myelodysplastic syndrome (MDS) is highly deregulated and presents promising avenues for understanding disease pathogenesis and potential molecular dependencies. Here, we evaluated the transcriptomic landscape in MDS in multiple independent studies focusing more on metabolomics pathways. Identifying molecular dependencies will pave the way for a more precise disease stratification as well as the development of novel personalized treatment strategies. The study adopted a retrospective, cross-sectional approach, utilizing transcriptomic data from multiple MDS studies. The transcriptomic data were then subjected to comprehensive analyses, including differential gene expression, gene enrichment analysis, gene co-expression analysis, protein-protein interaction analyses, and survival analyses. PSAT1 showed a significant upregulation profile in MDS patients. This observed upregulation is correlated with the deregulation of immune-related pathways in MDS samples. This observation suggests a novel role for PSAT1 in immune modulation and potentially in augmenting immune evasion, which may lead to poor prognosis. This was evident in other tumors in the TCGA database, where cancer patients with high PSAT1 expression have a shorter overall survival. This study unveils a novel potential therapeutic avenue in MDS. Identifying the role of the PSAT1 gene sheds light on the disease's intricate biology, highlighting the ongoing cross-talk between metabolism and immune regulation, which may pave the way for innovative treatment modalities.
Collapse
Affiliation(s)
- Sael Alatawi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Waseem Alzamzami
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| |
Collapse
|
17
|
Contento G, Wilson JAA, Selvarajah B, Platé M, Guillotin D, Morales V, Trevisani M, Pitozzi V, Bianchi K, Chambers RC. Pyruvate metabolism dictates fibroblast sensitivity to GLS1 inhibition during fibrogenesis. JCI Insight 2024; 9:e178453. [PMID: 39315548 PMCID: PMC11457851 DOI: 10.1172/jci.insight.178453] [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: 12/13/2023] [Accepted: 08/07/2024] [Indexed: 09/25/2024] Open
Abstract
Fibrosis is a chronic disease characterized by excessive extracellular matrix production, which leads to disruption of organ function. Fibroblasts are key effector cells of this process, responding chiefly to the pleiotropic cytokine transforming growth factor-β1 (TGF-β1), which promotes fibroblast to myofibroblast differentiation. We found that extracellular nutrient availability profoundly influenced the TGF-β1 transcriptome of primary human lung fibroblasts and that biosynthesis of amino acids emerged as a top enriched TGF-β1 transcriptional module. We subsequently uncovered a key role for pyruvate in influencing glutaminase (GLS1) inhibition during TGF-β1-induced fibrogenesis. In pyruvate-replete conditions, GLS1 inhibition was ineffective in blocking TGF-β1-induced fibrogenesis, as pyruvate can be used as the substrate for glutamate and alanine production via glutamate dehydrogenase (GDH) and glutamic-pyruvic transaminase 2 (GPT2), respectively. We further show that dual targeting of either GPT2 or GDH in combination with GLS1 inhibition was required to fully block TGF-β1-induced collagen synthesis. These findings embolden a therapeutic strategy aimed at additional targeting of mitochondrial pyruvate metabolism in the presence of a glutaminolysis inhibitor to interfere with the pathological deposition of collagen in the setting of pulmonary fibrosis and potentially other fibrotic conditions.
Collapse
Affiliation(s)
- Greg Contento
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London, United Kingdom
| | - Jo-Anne A.M. Wilson
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London, United Kingdom
| | - Brintha Selvarajah
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London, United Kingdom
| | - Manuela Platé
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London, United Kingdom
| | - Delphine Guillotin
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London, United Kingdom
| | - Valle Morales
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | | | - Vanessa Pitozzi
- Corporate Pre-Clinical R&D, Chiesi Farmaceutici S.p.A., Parma, Italy
| | - Katiuscia Bianchi
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | - Rachel C. Chambers
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London, United Kingdom
| |
Collapse
|
18
|
Qiu Y, Stamatatos OT, Hu Q, Ruiter Swain J, Russo S, Sann A, Costa ASH, Violante S, Spector DL, Cross JR, Lukey MJ. The unique catalytic properties of PSAT1 mediate metabolic adaptation to glutamine blockade. Nat Metab 2024; 6:1529-1548. [PMID: 39192144 PMCID: PMC11490312 DOI: 10.1038/s42255-024-01104-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 07/09/2024] [Indexed: 08/29/2024]
Abstract
Cultured cancer cells frequently rely on the consumption of glutamine and its subsequent hydrolysis by glutaminase (GLS). However, this metabolic addiction can be lost in the tumour microenvironment, rendering GLS inhibitors ineffective in the clinic. Here we show that glutamine-addicted breast cancer cells adapt to chronic glutamine starvation, or GLS inhibition, via AMPK-mediated upregulation of the serine synthesis pathway (SSP). In this context, the key product of the SSP is not serine, but α-ketoglutarate (α-KG). Mechanistically, we find that phosphoserine aminotransferase 1 (PSAT1) has a unique capacity for sustained α-KG production when glutamate is depleted. Breast cancer cells with resistance to glutamine starvation or GLS inhibition are highly dependent on SSP-supplied α-KG. Accordingly, inhibition of the SSP prevents adaptation to glutamine blockade, resulting in a potent drug synergism that suppresses breast tumour growth. These findings highlight how metabolic redundancy can be context dependent, with the catalytic properties of different metabolic enzymes that act on the same substrate determining which pathways can support tumour growth in a particular nutrient environment. This, in turn, has practical consequences for therapies targeting cancer metabolism.
Collapse
Affiliation(s)
- Yijian Qiu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Qingting Hu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY, USA
| | - Jed Ruiter Swain
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- CSHL School of Biological Sciences, Cold Spring Harbor, NY, USA
| | - Suzanne Russo
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ava Sann
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ana S H Costa
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Matterworks Inc., Somerville, MA, USA
| | - Sara Violante
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Justin R Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
19
|
Fuster-Martínez I, Calatayud S. The current landscape of antifibrotic therapy across different organs: A systematic approach. Pharmacol Res 2024; 205:107245. [PMID: 38821150 DOI: 10.1016/j.phrs.2024.107245] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Fibrosis is a common pathological process that can affect virtually all the organs, but there are hardly any effective therapeutic options. This has led to an intense search for antifibrotic therapies over the last decades, with a great number of clinical assays currently underway. We have systematically reviewed all current and recently finished clinical trials involved in the development of new antifibrotic drugs, and the preclinical studies analyzing the relevance of each of these pharmacological strategies in fibrotic processes affecting tissues beyond those being clinically studied. We analyze and discuss this information with the aim of determining the most promising options and the feasibility of extending their therapeutic value as antifibrotic agents to other fibrotic conditions.
Collapse
Affiliation(s)
- Isabel Fuster-Martínez
- Departamento de Farmacología, Universitat de València, Valencia 46010, Spain; FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia 46020, Spain.
| | - Sara Calatayud
- Departamento de Farmacología, Universitat de València, Valencia 46010, Spain; CIBERehd (Centro de Investigación Biomédica en Red - Enfermedades Hepáticas y Digestivas), Spain.
| |
Collapse
|
20
|
Shin KWD, Atalay MV, Cetin-Atalay R, O'Leary EM, Glass ME, Szafran JCH, Woods PS, Meliton AY, Shamaa OR, Tian Y, Mutlu GM, Hamanaka RB. ATF4 and mTOR regulate metabolic reprogramming in TGF-β-treated lung fibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598694. [PMID: 38915485 PMCID: PMC11195155 DOI: 10.1101/2024.06.12.598694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Idiopathic pulmonary fibrosis is a fatal disease characterized by the TGF-β-dependent activation of lung fibroblasts, leading to excessive deposition of collagen proteins and progressive replacement of healthy lung with scar tissue. We and others have shown that fibroblast activation is supported by metabolic reprogramming, including the upregulation of the de novo synthesis of glycine, the most abundant amino acid found in collagen protein. How fibroblast metabolic reprogramming is regulated downstream of TGF-β is incompletely understood. We and others have shown that TGF-β-mediated activation of the Mechanistic Target of Rapamycin Complex 1 (mTORC1) and downstream upregulation of Activating Transcription Factor 4 (ATF4) promote increased expression of the enzymes required for de novo glycine synthesis; however, whether mTOR and ATF4 regulate other metabolic pathways in lung fibroblasts has not been explored. Here, we used RNA sequencing to determine how both ATF4 and mTOR regulate gene expression in human lung fibroblasts following TGF-β. We found that ATF4 primarily regulates enzymes and transporters involved in amino acid homeostasis as well as aminoacyl-tRNA synthetases. mTOR inhibition resulted not only in the loss of ATF4 target gene expression, but also in the reduced expression of glycolytic enzymes and mitochondrial electron transport chain subunits. Analysis of TGF-β-induced changes in cellular metabolite levels confirmed that ATF4 regulates amino acid homeostasis in lung fibroblasts while mTOR also regulates glycolytic and TCA cycle metabolites. We further analyzed publicly available single cell RNAseq data sets and found increased expression of ATF4 and mTOR metabolic targets in pathologic fibroblast populations from the lungs of IPF patients. Our results provide insight into the mechanisms of metabolic reprogramming in lung fibroblasts and highlight novel ATF4 and mTOR-dependent pathways that may be targeted to inhibit fibrotic processes.
Collapse
Affiliation(s)
- Kun Woo D Shin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | | | - Rengul Cetin-Atalay
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Erin M O'Leary
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Mariel E Glass
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Jennifer C Houpy Szafran
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Parker S Woods
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Angelo Y Meliton
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Obada R Shamaa
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Yufeng Tian
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Gökhan M Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Robert B Hamanaka
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
21
|
Lin J, Hou L, Zhao X, Zhong J, Lv Y, Jiang X, Ye B, Qiao Y. Switch of ELF3 and ATF4 transcriptional axis programs the amino acid insufficiency-linked epithelial-to-mesenchymal transition. Mol Ther 2024; 32:1956-1969. [PMID: 38627967 PMCID: PMC11184330 DOI: 10.1016/j.ymthe.2024.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/19/2024] [Accepted: 04/12/2024] [Indexed: 04/29/2024] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) that endows cancer cells with increased invasive and migratory capacity enables cancer dissemination and metastasis. This process is tightly associated with metabolic reprogramming acquired for rewiring cell status and signaling pathways for survival in dietary insufficiency conditions. However, it remains largely unclear how transcription factor (TF)-mediated transcriptional programs are modulated during the EMT process. Here, we reveal that depletion of a key epithelial TF, ELF3 (E74-like factor-3), triggers a transforming growth factor β (TGF-β) signaling activation-like mesenchymal transcriptomic profile and metastatic features linked to the aminoacyl-tRNA biogenesis pathway. Moreover, the transcriptome alterations elicited by ELF3 depletion perfectly resemble an ATF4-dependent weak response to amino acid starvation. Intriguingly, we observe an exclusive enrichment of ELF3 and ATF4 in epithelial and TGF-β-induced or ELF3-depletion-elicited mesenchymal enhancers, respectively, with rare co-binding on altered enhancers. We also find that the upregulation of aminoacyl-tRNA synthetases and some mesenchymal genes upon amino acid deprivation is diminished in ATF4-depleted cells. In sum, the loss of ELF3 binding on epithelial enhancers and the gain of ATF4 binding on the enhancers of mesenchymal factors and amino acid deprivation responsive genes facilitate the loss of epithelial cell features and the gain of TGF-β-signaling-associated mesenchymal signatures, which further promote lung cancer cell metastasis.
Collapse
Affiliation(s)
- Jianxiang Lin
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China; Shanghai Institute of Precision Medicine, Shanghai 200125, China
| | - Linjun Hou
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xin Zhao
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Jingli Zhong
- College of Life Science, Guangzhou University, Guangzhou 510006, China
| | - Yilv Lv
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xiaohua Jiang
- Center for Reproduction and Genetics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China.
| | - Bo Ye
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Yunbo Qiao
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China; Shanghai Institute of Precision Medicine, Shanghai 200125, China.
| |
Collapse
|
22
|
Wang Y, Wang X, Du C, Wang Z, Wang J, Zhou N, Wang B, Tan K, Fan Y, Cao P. Glycolysis and beyond in glucose metabolism: exploring pulmonary fibrosis at the metabolic crossroads. Front Endocrinol (Lausanne) 2024; 15:1379521. [PMID: 38854692 PMCID: PMC11157045 DOI: 10.3389/fendo.2024.1379521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/07/2024] [Indexed: 06/11/2024] Open
Abstract
At present, pulmonary fibrosis (PF) is a prevalent and irreversible lung disease with limited treatment options, and idiopathic pulmonary fibrosis (IPF) is one of its most common forms. Recent research has highlighted PF as a metabolic-related disease, including dysregulated iron, mitochondria, lipid, and glucose homeostasis. Systematic reports on the regulatory roles of glucose metabolism in PF are rare. This study explores the intricate relationships and signaling pathways between glucose metabolic processes and PF, delving into how key factors involved in glucose metabolism regulate PF progression, and the interplay between them. Specifically, we examined various enzymes, such as hexokinase (HK), 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), pyruvate kinase (PK), and lactate dehydrogenase (LDH), illustrating their regulatory roles in PF. It highlights the significance of lactate, alongside the role of pyruvate dehydrogenase kinase (PDK) and glucose transporters (GLUTs) in modulating pulmonary fibrosis and glucose metabolism. Additionally, critical regulatory factors such as transforming growth factor-beta (TGF-β), interleukin-1 beta (IL-1β), and hypoxia-inducible factor 1 subunit alpha (HIF-1α) were discussed, demonstrating their impact on both PF and glucose metabolic pathways. It underscores the pivotal role of AMP-activated protein kinase (AMPK) in this interplay, drawing connections between diabetes mellitus, insulin, insulin-like growth factors, and peroxisome proliferator-activated receptor gamma (PPARγ) with PF. This study emphasizes the role of key enzymes, regulators, and glucose transporters in fibrogenesis, suggesting the potential of targeting glucose metabolism for the clinical diagnosis and treatment of PF, and proposing new promising avenues for future research and therapeutic development.
Collapse
Affiliation(s)
- Yuejiao Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| | - Xue Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| | - Chaoqi Du
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| | - Zeming Wang
- Department of Laboratory, Hebei Provincial People’s Hospital, Shijiazhuang, Hebei, China
| | - Jiahui Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| | - Nan Zhou
- Department of Gynecology, Xingtai People’s Hospital, Xingtai, Hebei, China
| | - Baohua Wang
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ke Tan
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| | - Yumei Fan
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| | - Pengxiu Cao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Collaborative Innovation Center for Eco-Environment, Shijiazhuang, Hebei, China
| |
Collapse
|
23
|
Li S, Ouyang X, Sun H, Jin J, Chen Y, Li L, Wang Q, He Y, Wang J, Chen T, Zhong Q, Liang Y, Pierre P, Zou Q, Ye Y, Su B. DEPDC5 protects CD8 + T cells from ferroptosis by limiting mTORC1-mediated purine catabolism. Cell Discov 2024; 10:53. [PMID: 38763950 PMCID: PMC11102918 DOI: 10.1038/s41421-024-00682-z] [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: 02/24/2024] [Accepted: 04/10/2024] [Indexed: 05/21/2024] Open
Abstract
Peripheral CD8+ T cell number is tightly controlled but the precise molecular mechanism regulating this process is still not fully understood. In this study, we found that epilepsy patients with loss of function mutation of DEPDC5 had reduced peripheral CD8+ T cells, and DEPDC5 expression positively correlated with tumor-infiltrating CD8+ T cells as well as overall cancer patient survival, indicating that DEPDC5 may control peripheral CD8+ T cell homeostasis. Significantly, mice with T cell-specific Depdc5 deletion also had reduced peripheral CD8+ T cells and impaired anti-tumor immunity. Mechanistically, Depdc5-deficient CD8+ T cells produced high levels of xanthine oxidase and lipid ROS due to hyper-mTORC1-induced expression of ATF4, leading to spontaneous ferroptosis. Together, our study links DEPDC5-mediated mTORC1 signaling with CD8+ T cell protection from ferroptosis, thereby revealing a novel strategy for enhancing anti-tumor immunity via suppression of ferroptosis.
Collapse
Affiliation(s)
- Song Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology at Basic Medical College, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology and Center for Immune-Related Diseases Research at Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinxing Ouyang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology at Basic Medical College, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Chest Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongxiang Sun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology at Basic Medical College, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology and Center for Immune-Related Diseases Research at Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingsi Jin
- Shanghai Institute of Immunology, Department of Immunology and Microbiology at Basic Medical College, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology at Basic Medical College, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology and Center for Immune-Related Diseases Research at Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology at Basic Medical College, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology and Center for Immune-Related Diseases Research at Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qijun Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology at Basic Medical College, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingzhong He
- Department of Neurology of Shanghai Children's Medical Center affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiwen Wang
- Department of Neurology of Shanghai Children's Medical Center affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tongxin Chen
- Department of Allergy and Immunology, Division of Immunology and Multidisciplinary Specialty Clinic, Institute of Pediatric Translational Medicine at Shanghai Children's Medical Center affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinming Liang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Philippe Pierre
- Shanghai Institute of Immunology, Department of Immunology and Microbiology at Basic Medical College, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, cedex 9, France
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Qiang Zou
- Shanghai Institute of Immunology, Department of Immunology and Microbiology at Basic Medical College, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youqiong Ye
- Shanghai Institute of Immunology, Department of Immunology and Microbiology at Basic Medical College, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology and Center for Immune-Related Diseases Research at Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Su
- Shanghai Institute of Immunology, Department of Immunology and Microbiology at Basic Medical College, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Gastroenterology and Center for Immune-Related Diseases Research at Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Key Laboratory of Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
24
|
Ziehr DR, Li F, Parnell KM, Krah NM, Leahy KJ, Guillermier C, Varon J, Baron RM, Maron BA, Philp NJ, Hariri LP, Kim EY, Steinhauser ML, Knipe RS, Rutter J, Oldham WM. Lactate transport inhibition therapeutically reprograms fibroblast metabolism in experimental pulmonary fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.591150. [PMID: 38712233 PMCID: PMC11071479 DOI: 10.1101/2024.04.25.591150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Myofibroblast differentiation, essential for driving extracellular matrix synthesis in pulmonary fibrosis, requires increased glycolysis. While glycolytic cells must export lactate, the contributions of lactate transporters to myofibroblast differentiation are unknown. In this study, we investigated how MCT1 and MCT4, key lactate transporters, influence myofibroblast differentiation and experimental pulmonary fibrosis. Our findings reveal that inhibiting MCT1 or MCT4 reduces TGFβ-stimulated pulmonary myofibroblast differentiation in vitro and decreases bleomycin-induced pulmonary fibrosis in vivo. Through comprehensive metabolic analyses, including bioenergetics, stable isotope tracing, metabolomics, and imaging mass spectrometry in both cells and mice, we demonstrate that inhibiting lactate transport enhances oxidative phosphorylation, reduces reactive oxygen species production, and diminishes glucose metabolite incorporation into fibrotic lung regions. Furthermore, we introduce VB253, a novel MCT4 inhibitor, which ameliorates pulmonary fibrosis in both young and aged mice, with comparable efficacy to established antifibrotic therapies. These results underscore the necessity of lactate transport for myofibroblast differentiation, identify MCT1 and MCT4 as promising pharmacologic targets in pulmonary fibrosis, and support further evaluation of lactate transport inhibitors for patients for whom limited therapeutic options currently exist.
Collapse
Affiliation(s)
- David R. Ziehr
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Department of Medicine, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Fei Li
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | | | - Nathan M. Krah
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
- Department of Biochemistry, University of Utah, Salt Lake City, UT
| | - Kevin J. Leahy
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Christelle Guillermier
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Jack Varon
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Rebecca M. Baron
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Bradley A. Maron
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
- University of Maryland Institute for Health Computing, Bethesda, MD
| | - Nancy J. Philp
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Lida P. Hariri
- Department of Medicine, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Edy Y. Kim
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Matthew L. Steinhauser
- Aging Institute, University of Pittsburgh, Pittsburgh, PA
- UPMC Heart and Vascular Institute, UPMC Presbyterian, Pittsburgh, PA
| | - Rachel S. Knipe
- Department of Medicine, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Jared Rutter
- Department of Biochemistry, University of Utah, Salt Lake City, UT
- Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - William M. Oldham
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
25
|
Wang Z, Yang M, Li S, Chi H, Wang J, Xiao C. [A transcriptomic analysis of correlation between mitochondrial function and energy metabolism remodeling in mice with myocardial fibrosis following myocardial infarction]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:666-674. [PMID: 38708499 DOI: 10.12122/j.issn.1673-4254.2024.04.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
OBJECTIVE To investigate the changes of mitochondrial respiratory function during myocardial fibrosis in mice with myocardial infarction (MI) and its correlation with the increase of glycolytic flux. METHODS Forty C57BL/6N mice were randomized into two equal groups to receive sham operation or ligation of the left anterior descending coronary artery to induce acute MI. At 28 days after the operation, 5 mice from each group were euthanized and left ventricular tissue samples were collected for transcriptomic sequencing. FPKM method was used to calculate gene expression levels to identify the differentially expressed genes (DEGs) in MI mice, which were analyzed using GO and KEGG databases to determine the pathways affecting the disease process. Heat maps were drawn to show the differential expressions of the pathways and the related genes in the enrichment analysis. In primary cultures of neonatal mouse cardiac fibroblasts (CFs), the changes in mitochondrial respiration and glycolysis levels in response to treatment with the pro-fibrotic agonist TGF-β1 were analyzed using Seahorse experiment. RESULTS The mouse models of MI showed significantly increased diastolic and systolic left ventricular diameter (P < 0.05) and decreased left ventricular ejection fraction (P < 0.0001). A total of 124 up-regulated and 106 down-regulated DEGs were identified in the myocardial tissues of MI mice, and GO and KEGG enrichment analysis showed that these DEGs were significantly enriched in fatty acid metabolism, organelles and other metabolic pathways and in the mitochondria. Heat maps revealed fatty acid beta oxidation, mitochondrial dysfunction and increased glycolysis levels in MI mice. In the primary culture of CFs, treatment with TGF-β1 significantly reduced the basal and maximum respiratory levels and increased the basal and maximum glycolysis levels (P < 0.0001). CONCLUSION During myocardial fibrosis, energy metabolism remodeling occurs in the CFs, manifested by lowered mitochondrial function and increased energy generation through glycolysis.
Collapse
Affiliation(s)
- Z Wang
- Chinese PLA Medical School, Beijing 100853, China
- Department of Cardiovascular Surgery, Sixth Medical Center of Chinese PLA General Hospital, Beijing 100037, China
| | - M Yang
- Department of Cardiovascular Surgery, Sixth Medical Center of Chinese PLA General Hospital, Beijing 100037, China
| | - S Li
- Department of Cardiovascular Surgery, First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - H Chi
- Department of Cardiovascular Surgery, Sixth Medical Center of Chinese PLA General Hospital, Beijing 100037, China
| | - J Wang
- Department of Cardiovascular Surgery, Sixth Medical Center of Chinese PLA General Hospital, Beijing 100037, China
| | - C Xiao
- Department of Cardiovascular Surgery, Sixth Medical Center of Chinese PLA General Hospital, Beijing 100037, China
| |
Collapse
|
26
|
Grmai L, Michaca M, Lackner E, Nampoothiri V P N, Vasudevan D. Integrated stress response signaling acts as a metabolic sensor in fat tissues to regulate oocyte maturation and ovulation. Cell Rep 2024; 43:113863. [PMID: 38457339 PMCID: PMC11077669 DOI: 10.1016/j.celrep.2024.113863] [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: 03/21/2023] [Revised: 11/23/2023] [Accepted: 02/08/2024] [Indexed: 03/10/2024] Open
Abstract
Reproduction is an energy-intensive process requiring systemic coordination. However, the inter-organ signaling mechanisms that relay nutrient status to modulate reproductive output are poorly understood. Here, we use Drosophila melanogaster as a model to establish the integrated stress response (ISR) transcription factor, Atf4, as a fat tissue metabolic sensor that instructs oogenesis. We demonstrate that Atf4 regulates lipase activity to mediate yolk lipoprotein synthesis in the fat body. Depletion of Atf4 in the fat body also blunts oogenesis recovery after amino acid deprivation and re-feeding, suggestive of a nutrient-sensing role for Atf4. We also discovered that Atf4 promotes secretion of a fat-body-derived neuropeptide, CNMamide, which modulates neural circuits that promote egg-laying behavior (ovulation). Thus, we posit that ISR signaling in fat tissue acts as a "metabolic sensor" that instructs female reproduction-directly by impacting yolk lipoprotein production and follicle maturation and systemically by regulating ovulation.
Collapse
Affiliation(s)
- Lydia Grmai
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Manuel Michaca
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Emily Lackner
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Deepika Vasudevan
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
27
|
Hoque MM, Gbadegoye JO, Hassan FO, Raafat A, Lebeche D. Cardiac fibrogenesis: an immuno-metabolic perspective. Front Physiol 2024; 15:1336551. [PMID: 38577624 PMCID: PMC10993884 DOI: 10.3389/fphys.2024.1336551] [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: 11/16/2023] [Accepted: 03/07/2024] [Indexed: 04/06/2024] Open
Abstract
Cardiac fibrosis is a major and complex pathophysiological process that ultimately culminates in cardiac dysfunction and heart failure. This phenomenon includes not only the replacement of the damaged tissue by a fibrotic scar produced by activated fibroblasts/myofibroblasts but also a spatiotemporal alteration of the structural, biochemical, and biomechanical parameters in the ventricular wall, eliciting a reactive remodeling process. Though mechanical stress, post-infarct homeostatic imbalances, and neurohormonal activation are classically attributed to cardiac fibrosis, emerging evidence that supports the roles of immune system modulation, inflammation, and metabolic dysregulation in the initiation and progression of cardiac fibrogenesis has been reported. Adaptive changes, immune cell phenoconversions, and metabolic shifts in the cardiac nonmyocyte population provide initial protection, but persistent altered metabolic demand eventually contributes to adverse remodeling of the heart. Altered energy metabolism, mitochondrial dysfunction, various immune cells, immune mediators, and cross-talks between the immune cells and cardiomyocytes play crucial roles in orchestrating the transdifferentiation of fibroblasts and ensuing fibrotic remodeling of the heart. Manipulation of the metabolic plasticity, fibroblast-myofibroblast transition, and modulation of the immune response may hold promise for favorably modulating the fibrotic response following different cardiovascular pathological processes. Although the immunologic and metabolic perspectives of fibrosis in the heart are being reported in the literature, they lack a comprehensive sketch bridging these two arenas and illustrating the synchrony between them. This review aims to provide a comprehensive overview of the intricate relationship between different cardiac immune cells and metabolic pathways as well as summarizes the current understanding of the involvement of immune-metabolic pathways in cardiac fibrosis and attempts to identify some of the previously unaddressed questions that require further investigation. Moreover, the potential therapeutic strategies and emerging pharmacological interventions, including immune and metabolic modulators, that show promise in preventing or attenuating cardiac fibrosis and restoring cardiac function will be discussed.
Collapse
Affiliation(s)
- Md Monirul Hoque
- Departments of Physiology, The University of Tennessee Health Science Center, Memphis, TN, United States
- College of Graduate Health Sciences, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Joy Olaoluwa Gbadegoye
- Departments of Physiology, The University of Tennessee Health Science Center, Memphis, TN, United States
- College of Graduate Health Sciences, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Fasilat Oluwakemi Hassan
- Departments of Physiology, The University of Tennessee Health Science Center, Memphis, TN, United States
- College of Graduate Health Sciences, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Amr Raafat
- Departments of Physiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Djamel Lebeche
- Departments of Physiology, The University of Tennessee Health Science Center, Memphis, TN, United States
- College of Graduate Health Sciences, The University of Tennessee Health Science Center, Memphis, TN, United States
- Medicine-Cardiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
- Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
28
|
Li Z, Zhao J, Wu Y, Fan S, Yuan H, Xia J, Hu L, Yang J, Liu J, Wu X, Lin R, Yang L. TRAF2 decrease promotes the TGF-β-mTORC1 signal in MAFLD-HCC through enhancing AXIN1-mediated Smad7 degradation. FASEB J 2024; 38:e23491. [PMID: 38363556 DOI: 10.1096/fj.202302307r] [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: 11/08/2023] [Revised: 01/13/2024] [Accepted: 02/01/2024] [Indexed: 02/17/2024]
Abstract
According to recent research, metabolic-associated fatty liver disease (MAFLD) has emerged as an important underlying etiology of hepatocellular carcinoma (HCC). However, the molecular mechanism of MAFLD-HCC is still unclear. Tumor necrosis factor receptor-associated factor 2 (TRAF2) is the key molecule to mediate the signal of inflammatory NF-κB pathway. This study aims to investigate the potential dysregulation of TRAF2 and its biological function in MAFLD-HCC. Huh7 TRAF2-/- demonstrated increased tumor formation ability compared to huh7 TRAF2+/+ when stimulated with transforming growth factor-β (TGF-β). The decisive role of TGF-β in the development of MAFLD-HCC was confirmed through the specific depletion of TGF-β receptor II gene in the hepatocytes (Tgfbr2ΔHep) of mice. In TRAF2-/- cells treated with TGF-β, both the glycolysis rate and lipid synthesis were enhanced. We proved the signal of the mechanistic target of rapamycin complex 1 (mTORC1) could be activated in the presence of TGF-β, and was enhanced in TRAF2-/- cells. The coimmunoprecipitation (co-IP) experiments revealed that TRAF2 fortified the Smurf2-mediated ubiquitination degradation of AXIN1. Hence, TRAF2 depletion resulted in increased Smad7 degradation induced by AXIN1, thus promoting the TGF-β signal. We also discovered that PLX-4720 could bind with AXIN1 and restrained the tumor proliferation of TRAF2-/- in mice fed with high-fat diet (HFD). Our findings indicate that TRAF2 plays a significant role in the pathogenesis of MAFLD-HCC. The reduction of TRAF2 expression leads to the enhancement of the TGF-β-mTORC1 pathway by facilitating AXIN1-mediated Smad7 degradation.
Collapse
Affiliation(s)
- Zhonglin Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinfang Zhao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya Wu
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Siyuan Fan
- Cardiovascular Medicine Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hang Yuan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Xia
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lilin Hu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingze Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiazheng Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Xuefeng Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Lin
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Kim J, Ryu G, Seo J, Go M, Kim G, Yi S, Kim S, Lee H, Lee JY, Kim HS, Park MC, Shin DH, Shim H, Kim W, Lee SY. 5-aminosalicylic acid suppresses osteoarthritis through the OSCAR-PPARγ axis. Nat Commun 2024; 15:1024. [PMID: 38310093 PMCID: PMC10838344 DOI: 10.1038/s41467-024-45174-6] [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: 04/20/2023] [Accepted: 01/16/2024] [Indexed: 02/05/2024] Open
Abstract
Osteoarthritis (OA) is a progressive and irreversible degenerative joint disease that is characterized by cartilage destruction, osteophyte formation, subchondral bone remodeling, and synovitis. Despite affecting millions of patients, effective and safe disease-modifying osteoarthritis drugs are lacking. Here we reveal an unexpected role for the small molecule 5-aminosalicylic acid (5-ASA), which is used as an anti-inflammatory drug in ulcerative colitis. We show that 5-ASA competes with extracellular-matrix collagen-II to bind to osteoclast-associated receptor (OSCAR) on chondrocytes. Intra-articular 5-ASA injections ameliorate OA generated by surgery-induced medial-meniscus destabilization in male mice. Significantly, this effect is also observed when 5-ASA was administered well after OA onset. Moreover, mice with DMM-induced OA that are treated with 5-ASA at weeks 8-11 and sacrificed at week 12 have thicker cartilage than untreated mice that were sacrificed at week 8. Mechanistically, 5-ASA reverses OSCAR-mediated transcriptional repression of PPARγ in articular chondrocytes, thereby suppressing COX-2-related inflammation. It also improves chondrogenesis, strongly downregulates ECM catabolism, and promotes ECM anabolism. Our results suggest that 5-ASA could serve as a DMOAD.
Collapse
Affiliation(s)
- Jihee Kim
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Republic of Korea
| | - Gina Ryu
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
| | - Jeongmin Seo
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
| | - Miyeon Go
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
| | - Gyungmin Kim
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
| | - Sol Yi
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
| | - Suwon Kim
- Department of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Hana Lee
- Department of Biomedical Engineering, Yonsei University, Wonju, Republic of Korea
| | - June-Yong Lee
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Han Sung Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, Republic of Korea
| | - Min-Chan Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dong Hae Shin
- Department of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Hyunbo Shim
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
| | - Wankyu Kim
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea
| | - Soo Young Lee
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea.
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Republic of Korea.
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Shan B, Zhou H, Guo C, Liu X, Wu M, Zhai R, Chen J. Tanshinone IIA ameliorates energy metabolism dysfunction of pulmonary fibrosis using 13C metabolic flux analysis. J Pharm Anal 2024; 14:244-258. [PMID: 38464785 PMCID: PMC10921327 DOI: 10.1016/j.jpha.2023.09.008] [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: 05/26/2023] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 03/12/2024] Open
Abstract
Evidence indicates that metabolic reprogramming characterized by the changes in cellular metabolic patterns contributes to the pathogenesis of pulmonary fibrosis (PF). It is considered as a promising therapeutic target anti-PF. The well-documented against PF properties of Tanshinone IIA (Tan IIA) have been primarily attributed to its antioxidant and anti-inflammatory potency. Emerging evidence suggests that Tan IIA may target energy metabolism pathways, including glycolysis and tricarboxylic acid (TCA) cycle. However, the detailed and advanced mechanisms underlying the anti-PF activities remain obscure. In this study, we applied [U-13C]-glucose metabolic flux analysis (MFA) to examine metabolism flux disruption and modulation nodes of Tan IIA in PF. We identified that Tan IIA inhibited the glycolysis and TCA flux, thereby suppressing the production of transforming growth factor-β1 (TGF-β1)-dependent extracellular matrix and the differentiation and proliferation of myofibroblasts in vitro. We further revealed that Tan IIA inhibited the expression of key metabolic enzyme hexokinase 2 (HK2) by inhibiting phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR)/hypoxia-inducible factor 1α (HIF-1α) pathway activities, which decreased the accumulation of abnormal metabolites. Notably, we demonstrated that Tan IIA inhibited ATP citrate lyase (ACLY) activity, which reduced the collagen synthesis pathway caused by cytosol citrate consumption. Further, these results were validated in a mouse model of bleomycin-induced PF. This study was novel in exploring the mechanism of the occurrence and development of Tan IIA in treating PF using 13C-MFA technology. It provided a novel understanding of the mechanism of Tan IIA against PF from the perspective of metabolic reprogramming.
Collapse
Affiliation(s)
- Baixi Shan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Haoyan Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Congying Guo
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiaolu Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Mingyu Wu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Rao Zhai
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jun Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
31
|
Amiri M, Kiniry SJ, Possemato AP, Mahmood N, Basiri T, Dufour CR, Tabatabaei N, Deng Q, Bellucci MA, Harwalkar K, Stokes MP, Giguère V, Kaufman RJ, Yamanaka Y, Baranov PV, Tahmasebi S, Sonenberg N. Impact of eIF2α phosphorylation on the translational landscape of mouse embryonic stem cells. Cell Rep 2024; 43:113615. [PMID: 38159280 PMCID: PMC10962698 DOI: 10.1016/j.celrep.2023.113615] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/24/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024] Open
Abstract
The integrated stress response (ISR) is critical for cell survival under stress. In response to diverse environmental cues, eIF2α becomes phosphorylated, engendering a dramatic change in mRNA translation. The activation of ISR plays a pivotal role in the early embryogenesis, but the eIF2-dependent translational landscape in pluripotent embryonic stem cells (ESCs) is largely unexplored. We employ a multi-omics approach consisting of ribosome profiling, proteomics, and metabolomics in wild-type (eIF2α+/+) and phosphorylation-deficient mutant eIF2α (eIF2αA/A) mouse ESCs (mESCs) to investigate phosphorylated (p)-eIF2α-dependent translational control of naive pluripotency. We show a transient increase in p-eIF2α in the naive epiblast layer of E4.5 embryos. Absence of eIF2α phosphorylation engenders an exit from naive pluripotency following 2i (two chemical inhibitors of MEK1/2 and GSK3α/β) withdrawal. p-eIF2α controls translation of mRNAs encoding proteins that govern pluripotency, chromatin organization, and glutathione synthesis. Thus, p-eIF2α acts as a key regulator of the naive pluripotency gene regulatory network.
Collapse
Affiliation(s)
- Mehdi Amiri
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Stephen J Kiniry
- School of Biochemistry and Cell Biology, University College Cork, T12 XF62 Cork, Ireland
| | | | - Niaz Mahmood
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Tayebeh Basiri
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Catherine R Dufour
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Negar Tabatabaei
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Qiyun Deng
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Michael A Bellucci
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Keerthana Harwalkar
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada
| | - Matthew P Stokes
- Cell Signaling Technology, Inc., 3 Trask Lane, Danvers, MA 01923, USA
| | - Vincent Giguère
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Randal J Kaufman
- Degenerative Diseases Program, Center for Genetic Disorders and Aging Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Yojiro Yamanaka
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada
| | - Pavel V Baranov
- School of Biochemistry and Cell Biology, University College Cork, T12 XF62 Cork, Ireland
| | - Soroush Tahmasebi
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| | - Nahum Sonenberg
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada.
| |
Collapse
|
32
|
Huang X, Yang X, Xiang L, Chen Y. Serine metabolism in macrophage polarization. Inflamm Res 2024; 73:83-98. [PMID: 38070057 DOI: 10.1007/s00011-023-01815-y] [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: 07/20/2023] [Revised: 10/09/2023] [Accepted: 11/06/2023] [Indexed: 01/10/2024] Open
Abstract
OBJECTIVE Emerging studies have revealed that macrophages possess different dependences on the uptake, synthesis, and metabolism of serine for their activation and functionalization, necessitating our insight into how serine availability and utilization impact macrophage activation and inflammatory responses. METHODS This article summarizes the reports published domestically and internationally about the serine uptake, synthesis, and metabolic flux by the macrophages polarizing with distinct stimuli and under different pathologic conditions, and particularly analyzes how altered serine metabolism rewires the metabolic behaviors of polarizing macrophages and their genetic and epigenetic reprogramming. RESULTS Macrophages dynamically change serine metabolism to orchestrate their anabolism, redox balance, mitochondrial function, epigenetics, and post-translation modification, and thus match the distinct needs for both classical and alternative activation. CONCLUSION Serine metabolism coordinates multiple metabolic pathways to tailor macrophage polarization and their responses to different pathogenic attacks and thus holds the potential as therapeutic target for types of acute and chronic inflammatory diseases.
Collapse
Affiliation(s)
| | - Xue Yang
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, 28 West Changsheng Road, Hengyang, 421001, Hunan, China
| | - Li Xiang
- Hengyang Medical School, Hengyang, China
| | - Yuping Chen
- Hengyang Medical School, Hengyang, China.
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, 28 West Changsheng Road, Hengyang, 421001, Hunan, China.
| |
Collapse
|
33
|
Yan P, Liu J, Li Z, Wang J, Zhu Z, Wang L, Yu G. Glycolysis Reprogramming in Idiopathic Pulmonary Fibrosis: Unveiling the Mystery of Lactate in the Lung. Int J Mol Sci 2023; 25:315. [PMID: 38203486 PMCID: PMC10779333 DOI: 10.3390/ijms25010315] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/17/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung disease characterized by excessive deposition of fibrotic connective tissue in the lungs. Emerging evidence suggests that metabolic alterations, particularly glycolysis reprogramming, play a crucial role in the pathogenesis of IPF. Lactate, once considered a metabolic waste product, is now recognized as a signaling molecule involved in various cellular processes. In the context of IPF, lactate has been shown to promote fibroblast activation, myofibroblast differentiation, and extracellular matrix remodeling. Furthermore, lactate can modulate immune responses and contribute to the pro-inflammatory microenvironment observed in IPF. In addition, lactate has been implicated in the crosstalk between different cell types involved in IPF; it can influence cell-cell communication, cytokine production, and the activation of profibrotic signaling pathways. This review aims to summarize the current research progress on the role of glycolytic reprogramming and lactate in IPF and its potential implications to clarify the role of lactate in IPF and to provide a reference and direction for future research. In conclusion, elucidating the intricate interplay between lactate metabolism and fibrotic processes may lead to the development of innovative therapeutic strategies for IPF.
Collapse
Affiliation(s)
| | | | | | | | | | - Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China; (P.Y.); (J.L.); (Z.L.); (J.W.); (Z.Z.)
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China; (P.Y.); (J.L.); (Z.L.); (J.W.); (Z.Z.)
| |
Collapse
|
34
|
Kalinin A, Zubkova E, Menshikov M. Integrated Stress Response (ISR) Pathway: Unraveling Its Role in Cellular Senescence. Int J Mol Sci 2023; 24:17423. [PMID: 38139251 PMCID: PMC10743681 DOI: 10.3390/ijms242417423] [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: 11/02/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Cellular senescence is a complex process characterized by irreversible cell cycle arrest. Senescent cells accumulate with age, promoting disease development, yet the absence of specific markers hampers the development of selective anti-senescence drugs. The integrated stress response (ISR), an evolutionarily highly conserved signaling network activated in response to stress, globally downregulates protein translation while initiating the translation of specific protein sets including transcription factors. We propose that ISR signaling plays a central role in controlling senescence, given that senescence is considered a form of cellular stress. Exploring the intricate relationship between the ISR pathway and cellular senescence, we emphasize its potential as a regulatory mechanism in senescence and cellular metabolism. The ISR emerges as a master regulator of cellular metabolism during stress, activating autophagy and the mitochondrial unfolded protein response, crucial for maintaining mitochondrial quality and efficiency. Our review comprehensively examines ISR molecular mechanisms, focusing on ATF4-interacting partners, ISR modulators, and their impact on senescence-related conditions. By shedding light on the intricate relationship between ISR and cellular senescence, we aim to inspire future research directions and advance the development of targeted anti-senescence therapies based on ISR modulation.
Collapse
Affiliation(s)
- Alexander Kalinin
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia; (A.K.); (E.Z.)
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Ekaterina Zubkova
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia; (A.K.); (E.Z.)
| | - Mikhail Menshikov
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia; (A.K.); (E.Z.)
| |
Collapse
|
35
|
O’Reilly S. Interleukin-11 and its eminent role in tissue fibrosis: a possible therapeutic target. Clin Exp Immunol 2023; 214:154-161. [PMID: 37724596 PMCID: PMC10714194 DOI: 10.1093/cei/uxad108] [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: 06/27/2023] [Revised: 08/31/2023] [Accepted: 09/15/2023] [Indexed: 09/21/2023] Open
Abstract
Interleukin-11 is a cytokine from the IL-6 family of cytokines that includes IL-6 and oncostatin-M. Initially described for its role in platelet generation, it is now appreciated that this cytokine has multiple functions. Recently it has been found that IL-11 is critical in fibrosis in multiple different organ systems and systemically as in the autoimmune disease systemic sclerosis. Animal models of fibrosis have determined that animals with IL-11 receptor deletions have retarded fibrosis and that in wild-type animals IL-11 is found at the organ of fibrosis. Recent evidence suggests that IL-11 may be a master regulator of fibrosis regardless of end target organ. With the development of neutralizing antibodies targeting the cytokine in pre-clinical models this could be a possible therapeutic, in a disease in which no specific therapies exist. This review appraises the evidence of the role of IL-11 in tissue fibrosis, its signalling properties, and therapeutic targeting. The review ends with an appraisal of indications for which IL-11 modulation is targeted.
Collapse
|
36
|
Petrova B, Maynard AG, Wang P, Kanarek N. Regulatory mechanisms of one-carbon metabolism enzymes. J Biol Chem 2023; 299:105457. [PMID: 37949226 PMCID: PMC10758965 DOI: 10.1016/j.jbc.2023.105457] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
One-carbon metabolism is a central metabolic pathway critical for the biosynthesis of several amino acids, methyl group donors, and nucleotides. The pathway mostly relies on the transfer of a carbon unit from the amino acid serine, through the cofactor folate (in its several forms), and to the ultimate carbon acceptors that include nucleotides and methyl groups used for methylation of proteins, RNA, and DNA. Nucleotides are required for DNA replication, DNA repair, gene expression, and protein translation, through ribosomal RNA. Therefore, the one-carbon metabolism pathway is essential for cell growth and function in all cells, but is specifically important for rapidly proliferating cells. The regulation of one-carbon metabolism is a critical aspect of the normal and pathological function of the pathway, such as in cancer, where hijacking these regulatory mechanisms feeds an increased need for nucleotides. One-carbon metabolism is regulated at several levels: via gene expression, posttranslational modification, subcellular compartmentalization, allosteric inhibition, and feedback regulation. In this review, we aim to inform the readers of relevant one-carbon metabolism regulation mechanisms and to bring forward the need to further study this aspect of one-carbon metabolism. The review aims to integrate two major aspects of cancer metabolism-signaling downstream of nutrient sensing and one-carbon metabolism, because while each of these is critical for the proliferation of cancerous cells, their integration is critical for comprehensive understating of cellular metabolism in transformed cells and can lead to clinically relevant insights.
Collapse
Affiliation(s)
- Boryana Petrova
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Adam G Maynard
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA; Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Peng Wang
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Naama Kanarek
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; The Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA.
| |
Collapse
|
37
|
Selvarajah B, Platé M, Chambers RC. Pulmonary fibrosis: Emerging diagnostic and therapeutic strategies. Mol Aspects Med 2023; 94:101227. [PMID: 38000335 DOI: 10.1016/j.mam.2023.101227] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023]
Abstract
Fibrosis is the concluding pathological outcome and major cause of morbidity and mortality in a number of common chronic inflammatory, immune-mediated and metabolic diseases. The progressive deposition of a collagen-rich extracellular matrix (ECM) represents the cornerstone of the fibrotic response and culminates in organ failure and premature death. Idiopathic pulmonary fibrosis (IPF) represents the most rapidly progressive and lethal of all fibrotic diseases with a dismal median survival of 3.5 years from diagnosis. Although the approval of the antifibrotic agents, pirfenidone and nintedanib, for the treatment of IPF signalled a watershed moment for the development of anti-fibrotic therapeutics, these agents slow but do not halt disease progression or improve quality of life. There therefore remains a pressing need for the development of effective therapeutic strategies. In this article, we review emerging therapeutic strategies for IPF as well as the pre-clinical and translational approaches that will underpin a greater understanding of the key pathomechanisms involved in order to transform the way we diagnose and treat pulmonary fibrosis.
Collapse
Affiliation(s)
- Brintha Selvarajah
- Oncogenes and Tumour Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Manuela Platé
- Department of Respiratory Medicine (UCL Respiratory), Division of Medicine, University College London, UK
| | - Rachel C Chambers
- Department of Respiratory Medicine (UCL Respiratory), Division of Medicine, University College London, UK.
| |
Collapse
|
38
|
McGill CJ, Ewald CY, Benayoun BA. Sex-dimorphic expression of extracellular matrix genes in mouse bone marrow neutrophils. PLoS One 2023; 18:e0294859. [PMID: 38032907 PMCID: PMC10688658 DOI: 10.1371/journal.pone.0294859] [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: 05/31/2023] [Accepted: 11/08/2023] [Indexed: 12/02/2023] Open
Abstract
The mammalian innate immune system is sex-dimorphic. Neutrophils are the most abundant leukocyte in humans and represent innate immunity's first line of defense. We previously found that primary mouse bone marrow neutrophils show widespread sex-dimorphism throughout life, including at the transcriptional level. Extracellular matrix [ECM]-related terms were observed among the top sex-dimorphic genes. Since the ECM is emerging as an important regulator of innate immune responses, we sought to further investigate the transcriptomic profile of primary mouse bone marrow neutrophils at both the bulk and single-cell level to understand how biological sex may influence ECM component expression in neutrophils throughout life. Here, using curated gene lists of ECM components and unbiased weighted gene co-expression network analysis [WGCNA], we find that multiple ECM-related gene sets show widespread female-bias in expression in primary mouse neutrophils. Since many immune-related diseases (e.g., rheumatoid arthritis) are more prevalent in females, our work may provide insights into the pathogenesis of sex-dimorphic inflammatory diseases.
Collapse
Affiliation(s)
- Cassandra J. McGill
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Collin Y. Ewald
- Laboratory of Extracellular Matrix Regeneration, Department of Health Sciences and Technology, Institute of Translational Medicine, Swiss Federal Institute of Technology (ETH Zürich), Schwerzenbach, Switzerland
| | - Bérénice A. Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
- Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, California, United States of America
- Biochemistry and Molecular Medicine Department, USC Keck School of Medicine, Los Angeles, California, United States of America
- USC Norris Comprehensive Cancer Center, Epigenetics and Gene Regulation, Los Angeles, California, United States of America
- USC Stem Cell Initiative, Los Angeles, California, United States of America
| |
Collapse
|
39
|
Zhan JH, Wei J, Liu L, Xu YT, Ji H, Wang CN, Liu YJ, Zhu XY. Investigation of a UPR-Related Gene Signature Identifies the Pro-Fibrotic Effects of Thrombospondin-1 by Activating CD47/ROS/Endoplasmic Reticulum Stress Pathway in Lung Fibroblasts. Antioxidants (Basel) 2023; 12:2024. [PMID: 38136144 PMCID: PMC10740656 DOI: 10.3390/antiox12122024] [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: 10/09/2023] [Revised: 11/17/2023] [Accepted: 11/19/2023] [Indexed: 12/24/2023] Open
Abstract
Unfolded protein response (UPR) signaling and endoplasmic reticulum (ER) stress have been linked to pulmonary fibrosis. However, the relationship between UPR status and pulmonary function and prognosis in idiopathic pulmonary fibrosis (IPF) patients remains largely unknown. Through a series of bioinformatics analyses, we established a correlation between UPR status and pulmonary function in IPF patients. Furthermore, thrombospondin-1 (TSP-1) was identified as a potential biomarker for prognostic evaluation in IPF patients. By utilizing both bulk RNA profiling and single-cell RNA sequencing data, we demonstrated the upregulation of TSP-1 in lung fibroblasts during pulmonary fibrosis. Gene set enrichment analysis (GSEA) results indicated a positive association between TSP-1 expression and gene sets related to the reactive oxygen species (ROS) pathway in lung fibroblasts. TSP-1 overexpression alone induced mild ER stress and pulmonary fibrosis, and it even exacerbated bleomycin-induced ER stress and pulmonary fibrosis. Mechanistically, TSP-1 promoted ER stress and fibroblast activation through CD47-dependent ROS production. Treatment with either TSP-1 inhibitor or CD47 inhibitor significantly attenuated BLM-induced ER stress and pulmonary fibrosis. Collectively, these findings suggest that the elevation of TSP-1 during pulmonary fibrosis is not merely a biomarker but likely plays a pathogenic role in the fibrotic changes in the lung.
Collapse
Affiliation(s)
- Jun-Hui Zhan
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
| | - Juan Wei
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
- School of Sports and Health, Nanjing Sport Institute, Nanjing 210014, China;
| | - Lin Liu
- School of Sports and Health, Nanjing Sport Institute, Nanjing 210014, China;
| | - Yi-Tong Xu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
| | - Hui Ji
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
| | - Chang-Nan Wang
- Department of Physiology, Navy Medical University, Shanghai 200433, China;
| | - Yu-Jian Liu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
| | - Xiao-Yan Zhu
- Department of Physiology, Navy Medical University, Shanghai 200433, China;
| |
Collapse
|
40
|
Meliton AY, Cetin-Atalay R, Tian Y, Szafran JCH, Shin KWD, Cho T, Sun KA, Woods PS, Shamaa OR, Chen B, Muir A, Mutlu GM, Hamanaka RB. Mitochondrial One-Carbon Metabolism is Required for TGF-β-Induced Glycine Synthesis and Collagen Protein Production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.07.566074. [PMID: 37986788 PMCID: PMC10659399 DOI: 10.1101/2023.11.07.566074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
A hallmark of Idiopathic Pulmonary Fibrosis is the TGF-β-dependent activation of lung fibroblasts, leading to excessive deposition of collagen proteins and progressive scarring. We have previously shown that synthesis of collagen by lung fibroblasts requires de novo synthesis of glycine, the most abundant amino acid in collagen protein. TGF-β upregulates the expression of the enzymes of the de novo serine/glycine synthesis pathway in lung fibroblasts through mTORC1 and ATF4-dependent transcriptional programs. SHMT2, the final enzyme of the de novo serine/glycine synthesis pathway, transfers a one-carbon unit from serine to tetrahydrofolate (THF), producing glycine and 5,10-methylene-THF (meTHF). meTHF is converted back to THF in the mitochondrial one-carbon (1C) pathway through the sequential actions of MTHFD2 (which converts meTHF to 10-formyl-THF), and either MTHFD1L, which produces formate, or ALDH1L2, which produces CO2. It is unknown how the mitochondrial 1C pathway contributes to glycine biosynthesis or collagen protein production in fibroblasts, or fibrosis in vivo. Here, we demonstrate that TGF-β induces the expression of MTHFD2, MTHFD1L, and ALDH1L2 in human lung fibroblasts. MTHFD2 expression was required for TGF-β-induced cellular glycine accumulation and collagen protein production. Combined knockdown of both MTHFD1L and ALDH1L2 also inhibited glycine accumulation and collagen protein production downstream of TGF-β; however knockdown of either protein alone had no inhibitory effect, suggesting that lung fibroblasts can utilize either enzyme to regenerate THF. Pharmacologic inhibition of MTHFD2 recapitulated the effects of MTHFD2 knockdown in lung fibroblasts and ameliorated fibrotic responses after intratracheal bleomycin instillation in vivo. Our results provide insight into the metabolic requirements of lung fibroblasts and provide support for continued development of MTHFD2 inhibitors for the treatment of IPF and other fibrotic diseases.
Collapse
Affiliation(s)
- Angelo Y Meliton
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Rengül Cetin-Atalay
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Yufeng Tian
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Jennifer C Houpy Szafran
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Kun Woo D Shin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Takugo Cho
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Kaitlyn A Sun
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Parker S Woods
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Obada R Shamaa
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Bohao Chen
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Alexander Muir
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637
| | - Gökhan M Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| | - Robert B Hamanaka
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
41
|
Yakupova E, Semenovich D, Abramicheva P, Zorova L, Pevzner I, Andrianova N, Popkov V, Manskikh V, Bocharnikov A, Voronina Y, Zorov D, Plotnikov E. Effects of caloric restriction and ketogenic diet on renal fibrosis after ischemia/reperfusion injury. Heliyon 2023; 9:e21003. [PMID: 37928038 PMCID: PMC10623167 DOI: 10.1016/j.heliyon.2023.e21003] [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: 05/10/2023] [Revised: 09/25/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023] Open
Abstract
The beneficial effects of caloric restriction (CR) and a ketogenic diet (KD) have been previously shown when performed prior to kidney injury. We investigated the effects of CR and KD on fibrosis development after unilateral kidney ischemia/reperfusion (UIR). Post-treatment with CR significantly (p < 0.05) affected blood glucose (2-fold decrease), ketone bodies (3-fold increase), lactate (1.5-fold decrease), and lipids (1.4-fold decrease). In the kidney, CR improved succinate dehydrogenase and malate dehydrogenase activity by 2-fold each, but worsened fibrosis progression. Similar results were shown for the KD, which restored the post-UIR impaired activities of succinate dehydrogenase, malate dehydrogenase, and α-ketoglutarate dehydrogenase (which was decreased 2-fold) but had no effect on fibrosis progression. Thus, our study shows that the use of CR or KD after UIR did not reduce the development of fibrosis, as shown by hydroxyproline content, western-blotting, and RT-PCR, whereas it caused significant metabolic changes in kidney tissue after UIR.
Collapse
Affiliation(s)
- E.I. Yakupova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| | - D.S. Semenovich
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| | - P.A. Abramicheva
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| | - L.D. Zorova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow 117997, Russia
| | - I.B. Pevzner
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow 117997, Russia
| | - N.V. Andrianova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| | - V.A. Popkov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow 117997, Russia
| | - V.N. Manskikh
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| | - A.D. Bocharnikov
- Sechenov First Moscow State Medical University, Moscow 119992, Russia
| | - Y.A. Voronina
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow 119234, Russia
- Laboratory of Cardiac Electrophysiology, National Medical Research Center for Cardiology, Moscow 121552, Russia
| | - D.B. Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow 117997, Russia
| | - E.Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Moscow 117997, Russia
| |
Collapse
|
42
|
Jeon KI, Kumar A, Callan CL, DeMagistris M, MacRae S, Nehrke K, Huxlin KR. Blocking Mitochondrial Pyruvate Transport Alters Corneal Myofibroblast Phenotype: A New Target for Treating Fibrosis. Invest Ophthalmol Vis Sci 2023; 64:36. [PMID: 37870848 PMCID: PMC10599161 DOI: 10.1167/iovs.64.13.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/21/2023] [Indexed: 10/24/2023] Open
Abstract
Purpose The purpose of this study was to critically test the hypothesis that mitochondrial pyruvate carrier (MPC) function is essential for maintenance of the corneal myofibroblast phenotype in vitro and in vivo. Methods Protein and mRNA for canonical profibrotic markers were assessed in cultured cat corneal myofibroblasts generated via transforming growth factor (TGF)-β1 stimulation and treated with either the thiazolidinedione (TZD) troglitazone or the MPC inhibitor alpha-cyano-beta-(1-phenylindol-3-yl) acrylate (UK-5099). RNA sequencing was used to gain insight into signaling modules related to instructive, permissive, or corollary changes in gene expression following treatment. A feline photorefractive keratectomy (PRK) model of corneal wounding was used to test the efficacy of topical troglitazone at reducing α-smooth muscle actin (SMA)-positive staining when applied 2 to 4 weeks postoperatively, during peak fibrosis. Results Troglitazone caused cultured myofibroblasts to adopt a fibroblast-like phenotype through a noncanonical, peroxisome proliferator-activated receptor (PPAR)-γ-independent mechanism. Direct MPC inhibition using UK-5099 recapitulated this effect, but classic inhibitors of oxidative phosphorylation (OXPHOS) did not. Gene Set Enrichment Analysis (GSEA) of RNA sequencing data converged on energy substrate utilization and the Mitochondrial Permeability Transition pore as key players in myofibroblast maintenance. Finally, troglitazone applied onto an established zone of active fibrosis post-PRK significantly reduced stromal α-SMA expression. Conclusions Our results provide empirical evidence that metabolic remodeling in myofibroblasts creates selective vulnerabilities beyond simply mitochondrial energy production, and that these are critical for maintenance of the myofibroblast phenotype. For the first time, we provide proof-of-concept data showing that this remodeling can be exploited to treat existing corneal fibrosis via inhibition of the MPC.
Collapse
Affiliation(s)
- Kye-Im Jeon
- Department of Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Ankita Kumar
- Department of Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Christine L Callan
- Department of Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Margaret DeMagistris
- Department of Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Scott MacRae
- Department of Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Keith Nehrke
- Department of Medicine-Nephrology Division, University of Rochester, Rochester, New York, United States
| | - Krystel R Huxlin
- Department of Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York, United States
| |
Collapse
|
43
|
Hanquier Z, Misra J, Baxter R, Maiers JL. Stress and Liver Fibrogenesis: Understanding the Role and Regulation of Stress Response Pathways in Hepatic Stellate Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1363-1376. [PMID: 37422148 PMCID: PMC10548279 DOI: 10.1016/j.ajpath.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/24/2023] [Accepted: 06/06/2023] [Indexed: 07/10/2023]
Abstract
Stress response pathways are crucial for cells to adapt to physiological and pathologic conditions. Increased transcription and translation in response to stimuli place a strain on the cell, necessitating increased amino acid supply, protein production and folding, and disposal of misfolded proteins. Stress response pathways, such as the unfolded protein response (UPR) and the integrated stress response (ISR), allow cells to adapt to stress and restore homeostasis; however, their role and regulation in pathologic conditions, such as hepatic fibrogenesis, are unclear. Liver injury promotes fibrogenesis through activation of hepatic stellate cells (HSCs), which produce and secrete fibrogenic proteins to promote tissue repair. This process is exacerbated in chronic liver disease, leading to fibrosis and, if unchecked, cirrhosis. Fibrogenic HSCs exhibit activation of both the UPR and ISR, due in part to increased transcriptional and translational demands, and these stress responses play important roles in fibrogenesis. Targeting these pathways to limit fibrogenesis or promote HSC apoptosis is a potential antifibrotic strategy, but it is limited by our lack of mechanistic understanding of how the UPR and ISR regulate HSC activation and fibrogenesis. This article explores the role of the UPR and ISR in the progression of fibrogenesis, and highlights areas that require further investigation to better understand how the UPR and ISR can be targeted to limit hepatic fibrosis progression.
Collapse
Affiliation(s)
- Zachary Hanquier
- Department of Molecular and Medical Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jagannath Misra
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Reese Baxter
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jessica L Maiers
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
44
|
Kim H, Jang Y, Ryu J, Seo D, Lee S, Choi S, Kim D, Moh S, Shin J. The Dipeptide Gly-Pro (GP), Derived from Hibiscus sabdariffa, Exhibits Potent Antifibrotic Effects by Regulating the TGF-β1-ATF4-Serine/Glycine Biosynthesis Pathway. Int J Mol Sci 2023; 24:13616. [PMID: 37686422 PMCID: PMC10487435 DOI: 10.3390/ijms241713616] [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: 07/25/2023] [Revised: 08/26/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
TGF-β1, a key fibrotic cytokine, enhances both the expression and translocation of the activating transcriptional factor 4 (ATF4) and activates the serine/glycine biosynthesis pathway, which is crucial for augmenting collagen production. Targeting the TGF-β1-ATF4-serine/glycine biosynthesis pathway might offer a promising therapeutic approach for fibrotic diseases. In this study, we aimed to identify a proline-containing dipeptide in Hibiscus sabdariffa plant cells that modulates collagen synthesis. We induced Hibiscus sabdariffa plant cells and screened for a proline-containing dipeptide that can suppress TGF-β1-induced collagen synthesis in fibroblasts. Analyses were conducted using LC-MS/MS, RT-qPCR, Western blot analysis, and immunocytochemistry. We identified Gly-Pro (GP) from the extract of Hibiscus sabdariffa plant cells as a dipeptide capable of suppressing TGF-β1-induced collagen production. GP inhibited the phosphorylation of Smad2/3 and reduced the expression of ATF4, which is upregulated by TGF-β1. Notably, GP also decreased the expression of enzymes involved in the serine/glycine biosynthesis and glucose metabolism pathways, such as PHGDH, PSAT1, PSPH, SHMT2, and SLC2A1. Our findings indicate that the peptide GP, derived from Hibiscus sabdariffa plant cells, exhibits potent anti-fibrotic effects, potentially through its regulation of the TGF-β1-ATF4-serine/glycine biosynthesis pathway.
Collapse
Affiliation(s)
- HaiVin Kim
- Department of Biomedical Science, College of Life Science, Graduate School, CHA University, Seongnam 13488, Republic of Korea; (H.K.); (Y.J.); (D.S.)
| | - YoungSu Jang
- Department of Biomedical Science, College of Life Science, Graduate School, CHA University, Seongnam 13488, Republic of Korea; (H.K.); (Y.J.); (D.S.)
| | - JaeSang Ryu
- Department of Dermatology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam 13496, Republic of Korea; (J.R.); (D.K.)
| | - DaHye Seo
- Department of Biomedical Science, College of Life Science, Graduate School, CHA University, Seongnam 13488, Republic of Korea; (H.K.); (Y.J.); (D.S.)
| | - Sak Lee
- Plant Cell Research Institute of BIO-FD&C Co., Ltd., Incheon 21990, Republic of Korea;
| | - SungSoo Choi
- Daesang Holdings, Jung-gu, Seoul 04513, Republic of Korea;
| | - DongHyun Kim
- Department of Dermatology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam 13496, Republic of Korea; (J.R.); (D.K.)
| | - SangHyun Moh
- Plant Cell Research Institute of BIO-FD&C Co., Ltd., Incheon 21990, Republic of Korea;
| | - JungU Shin
- Department of Dermatology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam 13496, Republic of Korea; (J.R.); (D.K.)
| |
Collapse
|
45
|
Duarte PRA, Franco RR, Vilela DD, Caixeta DC, de Souza AV, Deconte SR, Mendes-Rodrigues C, Fidale TM, Espindola FS, Teixeira RR, Resende ES. Effects of an L-Leucine-Rich Diet on Liver and Kidneys in a Doxorubicin Toxicity Model. Life (Basel) 2023; 13:1823. [PMID: 37763227 PMCID: PMC10532802 DOI: 10.3390/life13091823] [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: 06/28/2023] [Revised: 07/24/2023] [Accepted: 08/02/2023] [Indexed: 09/29/2023] Open
Abstract
Supplements and diets containing L-leucine, a branched-chain amino acid, have been considered beneficial for controlling oxidative stress and maintaining cardiac tissue in toxicity models using doxorubicin, a drug widely used in cancer treatment. However, there is a lack of studies in the literature that assess the effects of this diet on other organs and tissues, such as the liver and kidneys. Therefore, this study aimed to evaluate the effects of a leucine-rich diet on the liver and kidneys of healthy rats submitted to the doxorubicin toxicity model by analyzing biomarkers of oxidative stress and histological parameters. The animals were divided into four groups: naive, doxorubicin, L-leucine, and doxorubicin + L-leucine, and the diet was standardized with 5% L-leucine and a dose of 7.5 mg/kg of doxorubicin. We evaluated tissue injury parameters and biomarkers of oxidative stress, including enzymes, antioxidant profile, and oxidized molecules, in the liver and kidneys. Although some studies have indicated benefits of a diet rich in L-leucine for the muscle tissue of animals that received doxorubicin, our results showed that the liver was the most affected organ by the L-leucine-rich diet since the diet reduced its antioxidant defenses and increased the deposit of collagen and fat in the hepatic tissue. In the kidneys, the main alteration was the reduction in the number of glomeruli. These results contribute to the scientific literature and encourage further studies to evaluate the effects of an L-leucine-rich diet or its supplementation, alone or combined with doxorubicin using an animal model of cancer. Therefore, our study concludes that the leucine-rich diet itself was harmful and, when co-administered with doxorubicin, was not able to maintain the antioxidant defenses and tissue structure of the evaluated organs.
Collapse
Affiliation(s)
- Poliana Rodrigues Alves Duarte
- Faculdade de Medicina, Universidade Federal de Uberlândia, Uberlândia 38400-902, MG, Brazil; (P.R.A.D.); (S.R.D.); (C.M.-R.); (T.M.F.)
| | - Rodrigo Rodrigues Franco
- Departamento de Medicina, Universidade Federal de Catalão, Catalão 75706-881, GO, Brazil;
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia 38400-902, MG, Brazil; (D.D.V.); (D.C.C.); (A.V.d.S.)
| | - Danielle Diniz Vilela
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia 38400-902, MG, Brazil; (D.D.V.); (D.C.C.); (A.V.d.S.)
| | - Douglas Carvalho Caixeta
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia 38400-902, MG, Brazil; (D.D.V.); (D.C.C.); (A.V.d.S.)
| | - Adriele Vieira de Souza
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia 38400-902, MG, Brazil; (D.D.V.); (D.C.C.); (A.V.d.S.)
| | - Simone Ramos Deconte
- Faculdade de Medicina, Universidade Federal de Uberlândia, Uberlândia 38400-902, MG, Brazil; (P.R.A.D.); (S.R.D.); (C.M.-R.); (T.M.F.)
| | - Clesnan Mendes-Rodrigues
- Faculdade de Medicina, Universidade Federal de Uberlândia, Uberlândia 38400-902, MG, Brazil; (P.R.A.D.); (S.R.D.); (C.M.-R.); (T.M.F.)
| | - Thiago Montes Fidale
- Faculdade de Medicina, Universidade Federal de Uberlândia, Uberlândia 38400-902, MG, Brazil; (P.R.A.D.); (S.R.D.); (C.M.-R.); (T.M.F.)
- Departamento de Medicina, Universidade Federal de Catalão, Catalão 75706-881, GO, Brazil;
| | - Foued Salmen Espindola
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia 38400-902, MG, Brazil; (D.D.V.); (D.C.C.); (A.V.d.S.)
| | - Renata Roland Teixeira
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, Uberlândia 38400-902, MG, Brazil; (D.D.V.); (D.C.C.); (A.V.d.S.)
| | - Elmiro Santos Resende
- Faculdade de Medicina, Universidade Federal de Uberlândia, Uberlândia 38400-902, MG, Brazil; (P.R.A.D.); (S.R.D.); (C.M.-R.); (T.M.F.)
| |
Collapse
|
46
|
Hwang Y, Yun HJ, Jeong JW, Kim M, Joo S, Lee HK, Chang HS, Kim SM, Fang S. Co-inhibition of glutaminolysis and one-carbon metabolism promotes ROS accumulation leading to enhancement of chemotherapeutic efficacy in anaplastic thyroid cancer. Cell Death Dis 2023; 14:515. [PMID: 37573361 PMCID: PMC10423221 DOI: 10.1038/s41419-023-06041-2] [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: 03/03/2023] [Revised: 07/28/2023] [Accepted: 08/03/2023] [Indexed: 08/14/2023]
Abstract
Anaplastic thyroid cancer (ATC) is one of the most aggressive tumors with an extremely poor prognosis. Based on the several biological features related to glutamine metabolism in ATC, we hypothesized glutaminolysis inhibition induces cell death in ATC cells. However, glutamine metabolism inhibition triggered cell growth arrest independent of cell death in ATC, suggesting that other signaling pathways avoid glutamine metabolism inhibition-induced stress exist. To investigate the functional mechanism against glutamine metabolism inhibition, we conducted mRNA and ATAC-Sequencing data analysis and found that glutamine deprivation increased ATF4-mediated one-carbon metabolism. When we inhibited PHGDH, the first rate-limiting enzyme for one-carbon metabolism, cell growth arrest was promoted upon glutamine metabolism inhibition by accumulating intracellular ROS. We next observed that the co-inhibition of glutamine and one-carbon metabolism could augment the anticancer effects of drugs used in patients with ATC. Finally, single-cell RNA sequencing analysis revealed that one-carbon metabolism was strengthened through the evolutionary process from PTC to ATC. Collectively, our data demonstrate that one-carbon metabolism has a potential role of modulation of cell fate in metabolic stress and can be a therapeutic target for enhancing antitumor effects in ATC.
Collapse
Affiliation(s)
- Yeseong Hwang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Hyeok Jun Yun
- Department of Surgery, Thyroid Cancer Center, Institute of Refractory Thyroid Cancer, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Woong Jeong
- Department of Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Minki Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Seyeon Joo
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Hae-Kyung Lee
- Severance Biomedical Science Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hang-Seok Chang
- Department of Surgery, Thyroid Cancer Center, Institute of Refractory Thyroid Cancer, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seok-Mo Kim
- Department of Surgery, Thyroid Cancer Center, Institute of Refractory Thyroid Cancer, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| | - Sungsoon Fang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.
- Severance Biomedical Science Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Korea.
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
47
|
Phone Myint SMM, Sun LY. L-serine: Neurological Implications and Therapeutic Potential. Biomedicines 2023; 11:2117. [PMID: 37626614 PMCID: PMC10452085 DOI: 10.3390/biomedicines11082117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
L-serine is a non-essential amino acid that plays a vital role in protein synthesis, cell proliferation, development, and sphingolipid formation in the central nervous system. It exerts its effects through the activation of glycine receptors and upregulation of PPAR-γ, resulting in neurotransmitter synthesis, neuroprotection, and anti-inflammatory effects. L-serine shows potential as a protective agent in various neurological diseases and neurodegenerative disorders. Deficiency of L-serine and its downstream products has been linked to severe neurological deficits. Despite its crucial role, there is limited understanding of its mechanistic production and impact on glial and neuronal cells. Most of the focus has been on D-serine, the downstream product of L-serine, which has been implicated in a wide range of neurological diseases. However, L-serine is approved by FDA for supplemental use, while D-serine is not. Hence, it is imperative that we investigate the wider effects of L-serine, particularly in relation to the pathogenesis of several neurological deficits that, in turn, lead to diseases. This review aims to explore current knowledge surrounding L-serine and its potential as a treatment for various neurological diseases and neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Liou Y. Sun
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| |
Collapse
|
48
|
Ye MP, Lu WL, Rao QF, Li MJ, Hong HQ, Yang XY, Liu H, Kong JL, Guan RX, Huang Y, Hu QH, Wu FR. Mitochondrial stress induces hepatic stellate cell activation in response to the ATF4/TRIB3 pathway stimulation. J Gastroenterol 2023; 58:668-681. [PMID: 37150773 DOI: 10.1007/s00535-023-01996-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/19/2023] [Indexed: 05/09/2023]
Abstract
BACKGROUND The activation of hepatic stellate cells (HSCs) is the key step in the pathogenesis of liver fibrosis, which directly leads to fibrotic pathological changes in the hepatic tissue. Mitochondrial stress exacerbates inflammatory diseases by inducing pathogenic shifts in normal cells. However, the role of mitochondrial stress in HSC activation remains to be elucidated. METHODS: We analyzed the effect of mitochondrial stress on HSC activation. An in vivo hepatic fibrosis model was established by intraperitoneal injection of 40% carbon tetrachloride (CCl4) for 12 weeks. Additionally, using in vitro approach, HSC-T6 cells were treated with 10 ng/mL platelet-derived growth factor-BB (PDGF-BB) for 24 h. RESULTS Transcriptional activator 4 (ATF4) is highly expressed in fibrotic liver tissue samples and activated HSCs. We found that AAV8-shRNA-Atf4 alleviated liver fibrosis in rats. ATF4 promoted the activation of HSCs, which was induced by mitochondrial stress. The mechanisms involved ATF4 binding to a specific region of the tribble homologue 3 (TRIB3) promoter. Further, TRIB3 promoted HSCs activation mediated by mitochondrial stress. CONCLUSIONS ATF4 induces mitochondrial stress by upregulating TRIB3, leading to the activation of HSCs. Therefore, the inhibition of ATF4 during mitochondrial stress may be a promising therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Man-Ping Ye
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, People's Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Wei-Li Lu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, People's Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Qiu-Fan Rao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, People's Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Meng-Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, People's Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Hai-Qin Hong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, People's Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Xue-Ying Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, People's Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Hui Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, People's Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Jin-Ling Kong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, People's Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Ru-Xue Guan
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, People's Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, People's Republic of China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Qing-Hua Hu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Fan-Rong Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China.
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, People's Republic of China.
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, People's Republic of China.
| |
Collapse
|
49
|
Rajesh R, Atallah R, Bärnthaler T. Dysregulation of metabolic pathways in pulmonary fibrosis. Pharmacol Ther 2023; 246:108436. [PMID: 37150402 DOI: 10.1016/j.pharmthera.2023.108436] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/09/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive disorder of unknown origin and the most common interstitial lung disease. It progresses with the recruitment of fibroblasts and myofibroblasts that contribute to the accumulation of extracellular matrix (ECM) proteins, leading to the loss of compliance and alveolar integrity, compromising the gas exchange capacity of the lung. Moreover, while there are therapeutics available, they do not offer a cure. Thus, there is a pressing need to identify better therapeutic targets. With the advent of transcriptomics, proteomics, and metabolomics, the cellular mechanisms underlying disease progression are better understood. Metabolic homeostasis is one such factor and its dysregulation has been shown to impact the outcome of IPF. Several metabolic pathways involved in the metabolism of lipids, protein and carbohydrates have been implicated in IPF. While metabolites are crucial for the generation of energy, it is now appreciated that metabolites have several non-metabolic roles in regulating cellular processes such as proliferation, signaling, and death among several other functions. Through this review, we succinctly elucidate the role of several metabolic pathways in IPF. Moreover, we also discuss potential therapeutics which target metabolism or metabolic pathways.
Collapse
Affiliation(s)
- Rishi Rajesh
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Reham Atallah
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Thomas Bärnthaler
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria.
| |
Collapse
|
50
|
Samidurai A, Saravanan M, Ockaili R, Kraskauskas D, Lau SYV, Kodali V, Ramasamy S, Bhoopathi K, Nair M, Roh SK, Kukreja RC, Das A. Single-Dose Treatment with Rapamycin Preserves Post-Ischemic Cardiac Function through Attenuation of Fibrosis and Inflammation in Diabetic Rabbit. Int J Mol Sci 2023; 24:8998. [PMID: 37240345 PMCID: PMC10218967 DOI: 10.3390/ijms24108998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Robust activation of mTOR (mammalian target of rapamycin) signaling in diabetes exacerbates myocardial injury following lethal ischemia due to accelerated cardiomyocyte death with cardiac remodeling and inflammatory responses. We examined the effect of rapamycin (RAPA, mTOR inhibitor) on cardiac remodeling and inflammation following myocardial ischemia/reperfusion (I/R) injury in diabetic rabbits. Diabetic rabbits (DM) were subjected to 45 min of ischemia and 10 days of reperfusion by inflating/deflating a previously implanted hydraulic balloon occluder. RAPA (0.25 mg/kg, i.v.) or DMSO (vehicle) was infused 5 min before the onset of reperfusion. Post-I/R left ventricular (LV) function was assessed by echocardiography and fibrosis was evaluated by picrosirius red staining. Treatment with RAPA preserved LV ejection fraction and reduced fibrosis. Immunoblot and real-time PCR revealed that RAPA treatment inhibited several fibrosis markers (TGF-β, Galectin-3, MYH, p-SMAD). Furthermore, immunofluorescence staining revealed the attenuation of post-I/R NLRP3-inflammasome formation with RAPA treatment as shown by reduced aggregation of apoptosis speck-like protein with a caspase recruitment domain and active-form of caspase-1 in cardiomyocytes. In conclusion, our study suggests that acute reperfusion therapy with RAPA may be a viable strategy to preserve cardiac function with the alleviation of adverse post-infarct myocardial remodeling and inflammation in diabetic patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Rakesh C. Kukreja
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.)
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.)
| |
Collapse
|