1
|
Zhang X, Jiang A, An S, Guo C, You F, Huang Z, Feng S, Zhang Y, Chang X, Yang G, Meng X. Dietary resistant starch supplementation improves the fish growth, lipid metabolism and intestinal barrier in largemouth bass (Micropterus salmoides) fed high-fat diets. Int J Biol Macromol 2025; 306:141356. [PMID: 39988156 DOI: 10.1016/j.ijbiomac.2025.141356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Resistant starch (RS) is a novel type of prebiotic that exerts positive effects on lipid metabolism and intestinal flora. In this study, we investigated the effects of dietary RS on lipid metabolism and the intestinal barrier in largemouth bass (Micropterus salmoides). The experimental fish were fed either a control diet (C), a high-fat diet (H), or H diets supplemented with 0.5 %, 1.5 %, and 3 % RS (HRS0.5, HRS1.5, and HRS3.0). Dietary supplementation with 1.5 % and 3.0 % RS increased the final weight and feed utilization. Moreover, the hepatic crude protein content and the expression of genes related to lipid lipolysis were significantly higher in the HRS1.5 group compared to the H group, whereas hepatic crude lipid content and the expression of genes related to lipid synthesis were considerably lower in the HRS1.5 and HRS3.0 groups than in the H group. Additionally, hepatocyte vacuolation was alleviated in the HRS1.5 and HRS3.0 groups, and the number of liver lipid droplets was significantly decreased. Dietary supplementation with 1.5 % and 3.0 % RS downregulated the expression of pro-inflammatory factors while upregulating the expression of anti-inflammatory factors. Furthermore, analysis of gut microbiota composition revealed that RS supplementation increased the population of beneficial bacteria and short-chain fatty acid (SCFA) contents, decreased the abundance of pathogenic bacteria, and enhanced the diversity and richness of the intestinal flora. Non-targeted metabolomics analysis indicated that the levels of L-arginine and betaine were significantly higher in the HRS1.5 group, while levels of L-methionine and taurocholic acid were notably elevated in the HRS3.0 group. In conclusion, dietary supplementation with 1.5-3.0 % RS improved the balance of intestinal flora, promoted the growth of beneficial bacteria, adjusted the metabolites profile, and increased the SCFA levels. These results suggest that dietary supplementation with 1.5-3.0 % RS can restore the intestinal protective barrier, reduce hepatic lipid accumulation, and regulate lipid metabolism in largemouth bass.
Collapse
Affiliation(s)
- Xindang Zhang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, China
| | - Aixia Jiang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China
| | - Shuxia An
- College of Fisheries, Henan Normal University, Xinxiang 453007, China
| | - Chongchong Guo
- College of Fisheries, Henan Normal University, Xinxiang 453007, China
| | - Fu You
- College of Fisheries, Henan Normal University, Xinxiang 453007, China
| | - Zhenyi Huang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China
| | - Shikun Feng
- College of Fisheries, Henan Normal University, Xinxiang 453007, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, China
| | - Yanmin Zhang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, China
| | - Xulu Chang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, China
| | - Guokun Yang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, China
| | - Xiaolin Meng
- College of Fisheries, Henan Normal University, Xinxiang 453007, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, China.
| |
Collapse
|
2
|
Botella Martínez S, Varo Cenarruzabeitia N, Ribelles Segura MJ, Goñi Gironés E, Petrina Jáuregui E, Zugasti Murillo A, Marí Sanchís A, Redon Barragán H, Escuer Núñez I, Hernández Moreno A, Miranda Murúa C, Eguaras Córdoba I, Urribarri Marín AD. [Nutritional impact of beta-hydroxy-β-methylbutyrate supplementation in patients undergoing bariatric surgery]. NUTR HOSP 2025. [PMID: 40326309 DOI: 10.20960/nh.05569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025] Open
Abstract
INTRODUCTION bariatric surgery is an effective treatment but weight loss does not occur only at the expense of fat mass and preserving muscle mass is a priority. OBJECTIVE to evaluate the improvement in nutritional parameters of patients undergoing bariatric surgery after supplementation with beta-hydroxy-β-methylbutyrate (HMB). METHODOLOGY intervention study comparing 2 types of protein supplementation (with and without HMB) in the 3 months after surgery. All patients underwent a baseline and 3-month anthropometric, body composition, dynamometry and analytical study with usual nutritional determination, inflammation markers (CRP and IL-6), muscle markers (irisin, CK) and MRO (CTX, P1NP, sclerostin). Adherence was assessed and physical activity was recorded. RESULTS a total of 24 patients (87.5 % women) with a mean age of 52 ± 8 years were included. There were no statistically significant differences (p > 0.05) between intervention vs control in weight loss (17.5 ± 1.3 vs 19.1 ± 1.7 kg), MME (3.07 ± 1.7 vs 3.06 ± 136 kg), MG (12.9 ± 0.8 vs 14.7 ± 1.4 kg) or in dynamometry. Prealbumin levels in the intervention group were significantly higher (21 ± 1 vs 18 ± 0.8 mg/dl, p = 0.04) but there were no significant differences in the rest of the analytical markers. Significant changes in weight and fat mass were observed depending on physical activity. CONCLUSIONS after bariatric surgery, a significant weight reduction has been seen both at the expense of fat mass and lean mass in all patients. HMB supplementation has not shown statistically significant differences compared to the other protein supplementation to preserve muscle mass or function.
Collapse
|
3
|
Wang L, Shi F, Cao Y, Xie L. Multiple roles of branched-chain amino acid metabolism in tumour progression. J Biomed Sci 2025; 32:41. [PMID: 40205401 PMCID: PMC11983764 DOI: 10.1186/s12929-025-01132-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 03/09/2025] [Indexed: 04/11/2025] Open
Abstract
Metabolic reprogramming enables tumour cells to sustain their continuous proliferation and adapt to the ever-changing microenvironment. Branched-chain amino acids (BCAAs) and their metabolites are involved in intracellular protein synthesis and catabolism, signal transduction, epigenetic modifications, and the maintenance of oxidative homeostasis. Alterations in BCAA metabolism can influence the progression of various tumours. However, how BCAA metabolism is dysregulated differs among depending on tumour type; for example, it can manifest as decreased BCAA metabolism leading to BCAA accumulation, or as enhanced BCAA uptake and increased catabolism. In this review, we describe the role of BCAA metabolism in the progression of different tumours. As well as discuss how BCAA metabolic reprogramming drives tumour therapy resistance and evasion of the antitumour immune response, and how these pro-cancer effects are achieved in part by activating the mTORC signalling pathway. In-depth investigations into the potential mechanisms by which BCAA metabolic reprogramming affects tumorigenesis and tumour progression can enhance our understanding of the relationship between metabolism and cancer and provide new strategies for cancer therapy.
Collapse
Affiliation(s)
- Lin Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, XiangYa Hospital, Central South University, Changsha, 410078, China
| | - Feng Shi
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, XiangYa Hospital, Central South University, Changsha, 410078, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, XiangYa Hospital, Central South University, Changsha, 410078, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, 410078, China
| | - Longlong Xie
- Department of Radiology, The Affiliated Children's Hospital of Xiangya School of Medicine (Hunan Children's Hospital), Central South University, Changsha, 410078, China.
| |
Collapse
|
4
|
Xu J, Wang Y, Zhang J, Tang J, Zhou Z. The role of branched-chain amino acids in cardio-oncology: A review. Life Sci 2025; 372:123614. [PMID: 40189196 DOI: 10.1016/j.lfs.2025.123614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/18/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025]
Abstract
Cancer and cardiovascular diseases (CVDs) are global health challenges. In cancer patients, CVD is the second leading cause of death following disease progression. There are few specialized services for cardio-oncology patients worldwide currently. Branched-chain amino acids (BCAAs) are essential amino acids that promote protein synthesis and energy homeostasis. The disruption of BCAAs metabolism facilitates the development of cancer and CVDs while the benefit of BCAA supplement is full of controversy. In this review, we summarized BCAA-related studies in cardiometabolism, cancer and chemotherapy-induced cardiotoxicity, and provided our perspectives on the roles of BCAAs in cardio-oncology. We find that supplementation of BCAAs presents protective effects in cardiometabolic diseases, while the influence on cancer is intricate and varies across different types of cancers. Large-scale clinical studies are needed to understand the long-term effects of BCAA intake and its impact on different stages of the disease. BCAAs have potential to mitigate chemotherapy-induced cardiotoxicity. Continued research is still essential to understand the precise mechanisms, determine optimal dosage and timing, and assess the effectiveness of BCAA supplement in cardio-oncology, in particular clinical research.
Collapse
Affiliation(s)
- Jiaqi Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Cardiology, The First Hospital of Hebei Medical University, Hebei, China
| | - Jing Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Jingyi Tang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zhongyan Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong.
| |
Collapse
|
5
|
Dong J, Chen M, van Weering JRT, Li KW, Smit AB, Toonen RF, Verhage M. Rab10 regulates neuropeptide release by maintaining Ca 2+ homeostasis and protein synthesis. eLife 2025; 13:RP94930. [PMID: 40172954 PMCID: PMC11964448 DOI: 10.7554/elife.94930] [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] [Indexed: 04/04/2025] Open
Abstract
Dense core vesicles (DCVs) transport and release various neuropeptides and neurotrophins that control diverse brain functions, but the DCV secretory pathway remains poorly understood. Here, we tested a prediction emerging from invertebrate studies about the crucial role of the intracellular trafficking GTPase Rab10, by assessing DCV exocytosis at single-cell resolution upon acute Rab10 depletion in mature mouse hippocampal neurons, to circumvent potential confounding effects of Rab10's established role in neurite outgrowth. We observed a significant inhibition of DCV exocytosis in Rab10-depleted neurons, whereas synaptic vesicle exocytosis was unaffected. However, rather than a direct involvement in DCV trafficking, this effect was attributed to two ER-dependent processes, ER-regulated intracellular Ca2+ dynamics, and protein synthesis. Gene Ontology analysis of differentially expressed proteins upon Rab10 depletion identified substantial alterations in synaptic and ER/ribosomal proteins, including the Ca2+ pump SERCA2. In addition, ER morphology and dynamics were altered, ER Ca2+ levels were depleted, and Ca2+ homeostasis was impaired in Rab10-depleted neurons. However, Ca2+ entry using a Ca2+ ionophore still triggered less DCV exocytosis. Instead, leucine supplementation, which enhances protein synthesis, largely rescued DCV exocytosis deficiency. We conclude that Rab10 is required for neuropeptide release by maintaining Ca2+ dynamics and regulating protein synthesis. Furthermore, DCV exocytosis appeared more dependent on (acute) protein synthesis than synaptic vesicle exocytosis.
Collapse
Affiliation(s)
- Jian Dong
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - Mian Chen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - Jan RT van Weering
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), University Medical Center AmsterdamAmsterdamNetherlands
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), University Medical Center AmsterdamAmsterdamNetherlands
| |
Collapse
|
6
|
Tomar MS, Mohit, Kumar A, Shrivastava A. Circadian immunometabolism: A future insight for targeted therapy in cancer. Sleep Med Rev 2025; 80:102031. [PMID: 39603026 DOI: 10.1016/j.smrv.2024.102031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024]
Abstract
Circadian rhythms send messages to regulate the sleep-wake cycle in living beings, which, regulate various biological activities. It is well known that altered sleep-wake cycles affect host metabolism and significantly deregulate the host immunity. The dysregulation of circadian-related genes is critical for various malignancies. One of the hallmarks of cancer is altered metabolism, the effects of which spill into surrounding microenvironments. Here, we review the emerging literature linking the circadian immunometabolic axis to cancer. Small metabolites are the products of various metabolic pathways, that are usually perturbed in cancer. Genes that regulate circadian rhythms also regulate host metabolism and control metabolite content in the tumor microenvironment. Immune cell infiltration into the tumor site is critical to perform anticancer functions, and altered metabolite content affects their trafficking to the tumor site. A compromised immune response in the tumor microenvironment aids cancer cell proliferation and immune evasion, resulting in metastases. The role of circadian rhythms in these processes is largely overlooked and demands renewed attention in the search for targets against cancer growth and spread. The precision medicine approach requires targeting the circadian immune metabolism in cancer.
Collapse
Affiliation(s)
- Manendra Singh Tomar
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India
| | - Mohit
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India; Department of Prosthodontics, Faculty of Dental Sciences, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal, Madhya Pradesh, 462020, India.
| | - Ashutosh Shrivastava
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India.
| |
Collapse
|
7
|
Best L, Dost T, Esser D, Flor S, Gamarra AM, Haase M, Kadibalban AS, Marinos G, Walker A, Zimmermann J, Simon R, Schmidt S, Taubenheim J, Künzel S, Häsler R, Franzenburg S, Groth M, Waschina S, Rosenstiel P, Sommer F, Witte OW, Schmitt-Kopplin P, Baines JF, Frahm C, Kaleta C. Metabolic modelling reveals the aging-associated decline of host-microbiome metabolic interactions in mice. Nat Microbiol 2025; 10:973-991. [PMID: 40140706 PMCID: PMC11964932 DOI: 10.1038/s41564-025-01959-z] [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: 05/03/2024] [Accepted: 02/14/2025] [Indexed: 03/28/2025]
Abstract
Aging is accompanied by considerable changes in the gut microbiome, yet the molecular mechanisms driving aging and the role of the microbiome remain unclear. Here we combined metagenomics, transcriptomics and metabolomics from aging mice with metabolic modelling to characterize host-microbiome interactions during aging. Reconstructing integrated metabolic models of host and 181 mouse gut microorganisms, we show a complex dependency of host metabolism on known and previously undescribed microbial interactions. We observed a pronounced reduction in metabolic activity within the aging microbiome accompanied by reduced beneficial interactions between bacterial species. These changes coincided with increased systemic inflammation and the downregulation of essential host pathways, particularly in nucleotide metabolism, predicted to rely on the microbiota and critical for preserving intestinal barrier function, cellular replication and homeostasis. Our results elucidate microbiome-host interactions that potentially influence host aging processes. These pathways could serve as future targets for the development of microbiome-based anti-aging therapies.
Collapse
Affiliation(s)
- Lena Best
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thomas Dost
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Daniela Esser
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel/Lübeck, Germany
| | - Stefano Flor
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Andy Mercado Gamarra
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Madlen Haase
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - A Samer Kadibalban
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Georgios Marinos
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
- CAU Innovation GmbH, Kiel University, Kiel, Germany
| | - Alesia Walker
- Research Unit Analytical BioGeoChemistry, Helmholtz Munich, Neuherberg, Germany
| | - Johannes Zimmermann
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
- Evolutionary Ecology and Genetics, Zoological Institute, Kiel University, Kiel, Germany
- Antibiotic resistance group, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Rowena Simon
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Silvio Schmidt
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Jan Taubenheim
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sven Künzel
- Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Robert Häsler
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sören Franzenburg
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Marco Groth
- Core Facility Next-Generation Sequencing, Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Silvio Waschina
- Nutriinformatics, Institute of Human Nutrition and Food Science, Kiel University, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Felix Sommer
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Otto W Witte
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry, Helmholtz Munich, Neuherberg, Germany
- Institute of Analytical Food Chemistry, Technical University München, Freising, Germany
| | - John F Baines
- Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute of Experimental Medicine, Kiel University, Kiel, Germany
| | - Christiane Frahm
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Christoph Kaleta
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
8
|
Faiad J, Andrade MF, de Castro G, de Resende J, Coêlho M, Aquino G, Seelaender M. Muscle loss in cancer cachexia: what is the basis for nutritional support? Front Pharmacol 2025; 16:1519278. [PMID: 40078277 PMCID: PMC11897308 DOI: 10.3389/fphar.2025.1519278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/27/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer cachexia (CC) is characterized by significant skeletal muscle wasting, and contributes to diminished quality of life, while being associated with poorer response to treatment and with reduced survival. Chronic inflammation plays a central role in driving CC progression, within a complex interplay favoring catabolism. Although cachexia cannot be fully reversed by conventional nutritional support, nutritional intervention shows promise for the prevention and treatment of the syndrome. Of special interest are nutrients with antioxidant and anti-inflammatory potential and those that activate pathways involved in muscle mass synthesis and/or in the inhibition of muscle wasting. Extensive research has been carried out on novel nutritional supplements' power to mitigate CC impact, while the mechanisms through which some nutrients or bioactive compounds exert beneficial effects on muscle mass are still not totally clear. Here, we discuss the most studied supplements and nutritional strategies for dealing with muscle loss in CC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marilia Seelaender
- Cancer Metabolism Research Group, Faculdade de Medicina da Universidade de São Paulo, Departamento de Cirurgia, LIM 26-HC-USP, São Paulo, Brazil
| |
Collapse
|
9
|
Mone K, Garcia EJT, Abdullatif F, Rasquinha MT, Sur M, Hanafy M, Zinniel DK, Singh S, Thomas R, Barletta RG, Gebregiworgis T, Reddy J. Metabolic Reprogramming in Response to Freund's Adjuvants: Insights from Serum Metabolomics. Microorganisms 2025; 13:492. [PMID: 40142385 PMCID: PMC11944801 DOI: 10.3390/microorganisms13030492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
Freund's adjuvants have been used in vaccine and autoimmune settings, and their effects can be overlapping or unique to each. While both incomplete Freund's adjuvants (IFA) and complete Freund's adjuvants (CFA) influence antibody and T cell responses, the robust T helper 1 cytokines induced by the mycobacterial components make CFA the powerful immunostimulating adjuvant. In these studies, the adjuvant effects are investigated in a select population of cells, and the changes, if any, with the metabolic alterations in the systemic compartment are unclear. We investigated whether the effects of IFA and CFA can be influenced by the metabolic shifts in mice immunized with saline, IFA, or CFA using Mycobacterium tuberculosis var. bovis Bacillus Calmette-Guérin (BCG) as a positive control. After seven days of immunization, we analyzed the serum metabolite profiles using liquid chromatography coupled with high-resolution mass spectrometry and multivariate statistical analysis to identify metabolic features between the groups. The data revealed that, in the scores space, the CFA and BCG groups were more closely aligned compared to the saline group, while the IFA group displayed an intermediate profile. Furthermore, comparisons between the CFA and BCG groups showed more significant perturbations in lipid and amino acid metabolism, particularly involving glycerophospholipids, cysteine, and aromatic amino acids. In contrast, comparisons between the BCG and IFA groups indicated a more pronounced disruption in central energy metabolism pathways, such as the citric acid cycle and pyruvate metabolism. Together, the data suggest that the serum metabolite profiles in response to IFA and CFA might play a role in modulating the immune responses.
Collapse
Affiliation(s)
- Kiruthiga Mone
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.M.); (M.T.R.); (M.S.); (M.H.); (D.K.Z.); (S.S.); (R.G.B.)
| | - Eloy Jose Torres Garcia
- Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (E.J.T.G.); (F.A.)
| | - Fatema Abdullatif
- Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (E.J.T.G.); (F.A.)
| | - Mahima T. Rasquinha
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.M.); (M.T.R.); (M.S.); (M.H.); (D.K.Z.); (S.S.); (R.G.B.)
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Meghna Sur
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.M.); (M.T.R.); (M.S.); (M.H.); (D.K.Z.); (S.S.); (R.G.B.)
| | - Mostafa Hanafy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.M.); (M.T.R.); (M.S.); (M.H.); (D.K.Z.); (S.S.); (R.G.B.)
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Denise K. Zinniel
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.M.); (M.T.R.); (M.S.); (M.H.); (D.K.Z.); (S.S.); (R.G.B.)
| | - Shraddha Singh
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.M.); (M.T.R.); (M.S.); (M.H.); (D.K.Z.); (S.S.); (R.G.B.)
| | - Raymond Thomas
- Biotron Experimental Climate Change Research Centre, Department of Biology, Faculty of Science, University of Western Ontario, London, ON N6A 5B7, Canada;
| | - Raul G. Barletta
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.M.); (M.T.R.); (M.S.); (M.H.); (D.K.Z.); (S.S.); (R.G.B.)
| | - Teklab Gebregiworgis
- Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (E.J.T.G.); (F.A.)
- Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5W9, Canada
| | - Jay Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.M.); (M.T.R.); (M.S.); (M.H.); (D.K.Z.); (S.S.); (R.G.B.)
| |
Collapse
|
10
|
Tosello V, Di Martino L, Papathanassiu AE, Santa SD, Pizzi M, Mussolin L, Liu J, Van Vlierberghe P, Piovan E. BCAT1 is a NOTCH1 target and sustains the oncogenic function of NOTCH1. Haematologica 2025; 110:350-367. [PMID: 39234857 PMCID: PMC11788623 DOI: 10.3324/haematol.2024.285552] [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: 03/29/2024] [Accepted: 08/23/2024] [Indexed: 09/06/2024] Open
Abstract
High levels of branched-chain amino acid (BCAA) transaminase 1 (BCAT1) have been associated with tumor aggressiveness and drug resistance in several cancer types. Nevertheless, the mechanistic role of BCAT1 in T-cell acute lymphoblastic leukemia (T-ALL) remains uncertain. We provide evidence that Bcat1 was over-expressed following NOTCH1-induced transformation of leukemic progenitors and that NOTCH1 directly controlled BCAT1 expression by binding to a BCAT1 promoter. Further, using a NOTCH1 gain-of-function retroviral model of T-ALL, mouse cells genetically deficient for Bcat1 showed defects in developing leukemia. In murine T-ALL cells, Bcat1 depletion or inhibition redirected leucine metabolism towards production of 3-hydroxy butyrate (3-HB), an endogenous histone deacetylase inhibitor. Consistently, BCAT1-depleted cells showed altered protein acetylation levels which correlated with a pronounced sensitivity to DNA damaging agents. In human NOTCH1-dependent leukemias, high expression levels of BCAT1 may predispose to worse prognosis. Therapeutically, BCAT1 inhibition specifically synergized with etoposide to eliminate tumors in patient-derived xenograft models suggesting that BCAT1 inhibitors may have a part to play in salvage protocols for refractory T-ALL.
Collapse
Affiliation(s)
- Valeria Tosello
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua
| | | | | | - Silvia Dalla Santa
- Department of Surgery, Oncology and Gastroenterology, University of Padua
| | - Marco Pizzi
- Surgical Pathology and Cytopathology Unit, Department of Medicine - DIMED, University of Padua
| | - Lara Mussolin
- Unit of Onco-hematology, stem cell transplant and gene therapy, Department of Women's and Children's Health, University of Padua
| | - Jingjing Liu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN
| | | | - Erich Piovan
- Department of Surgery, Oncology and Gastroenterology, University of Padua; Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua.
| |
Collapse
|
11
|
Luo S, Yin J, Zhang J, Li P, Wen T, Li K, Tang J, Wang X, Li A, Chen L. Genetically Predicted Leucine Level Mediates Association Between CD4/CD8br T Lymphocytes and Insomnia. Cell Mol Neurobiol 2025; 45:15. [PMID: 39841266 PMCID: PMC11754360 DOI: 10.1007/s10571-025-01533-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025]
Abstract
Immune and metabolic factors play an important role in the onset and development of insomnia. This study aimed to investigate the causal relationship between insomnia and immune cells and metabolites. Data for 731 immune cell phenotypes, 1400 metabolites, and insomnia in this study were obtained from the GWAS open-access database. Two-way Mendelian randomization was used to (1) detect the causal relationship between immune cells and insomnia and (2) identify potential mediating metabolites. Mendelian randomization analysis identified eight immune cell phenotypes with a causal relationship to insomnia, and two immune cell phenotypes were protective factors for insomnia, namely CD8br %T cells and CD80 on CD62L + myeloid dendritic cells. The other six immune cell phenotypes were risk factors for insomnia, i.e., CD4/CD8br, CD16-CD56 on NKT, CCR2 on myeloid dendritic cells, CD40 on monocytes, CD38 on CD3-CD19-, and CD25 on CD45RA + CD4 not Treg. Further Mendelian randomization revealed 11 metabolites that were causally related to insomnia. Five metabolites were protective factors for insomnia, i.e., 3-hydroxy-3-methylglutarate, cholate, dodecanedioate, N-formylmethionine, and x-26054. Six metabolites were risk factors for insomnia, 3-amino-2-piperidone, 6-oxopiperdine-2-carboxylate, caffeine to theophylline ratio, leucine, maltose, and x-24736. In addition, our analysis showed that leucine mediated the association between CD4/CD8br and insomnia. From genetic information, we confirmed the causal relationship between insomnia, eight immune cell phenotypes, and eleven metabolite levels. Notably, we found a relationship between leucine-mediated CD4/CD8br and insomnia, providing evidence supporting the causal relationship between immune cell and insomnia, with plasma metabolites serving as mediators.
Collapse
Affiliation(s)
- Sumei Luo
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China
| | - Jianyin Yin
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China
| | - Jie Zhang
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China
| | - Pan Li
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China
| | - Tao Wen
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China
| | - Ke Li
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China
| | - Jing Tang
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China
| | - Xiaohong Wang
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China
| | - Aiyuan Li
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China
| | - Liang Chen
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China.
| |
Collapse
|
12
|
Lin DW, Zhang L, Zhang J, Chandrasekaran S. Inferring metabolic objectives and trade-offs in single cells during embryogenesis. Cell Syst 2025; 16:101164. [PMID: 39778581 PMCID: PMC11738665 DOI: 10.1016/j.cels.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/21/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025]
Abstract
While proliferating cells optimize their metabolism to produce biomass, the metabolic objectives of cells that perform non-proliferative tasks are unclear. The opposing requirements for optimizing each objective result in a trade-off that forces single cells to prioritize their metabolic needs and optimally allocate limited resources. Here, we present single-cell optimization objective and trade-off inference (SCOOTI), which infers metabolic objectives and trade-offs in biological systems by integrating bulk and single-cell omics data, using metabolic modeling and machine learning. We validated SCOOTI by identifying essential genes from CRISPR-Cas9 screens in embryonic stem cells, and by inferring the metabolic objectives of quiescent cells, during different cell-cycle phases. Applying this to embryonic cell states, we observed a decrease in metabolic entropy upon development. We further uncovered a trade-off between glutathione and biosynthetic precursors in one-cell zygote, two-cell embryo, and blastocyst cells, potentially representing a trade-off between pluripotency and proliferation. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Da-Wei Lin
- Center for Bioinformatics and Computational Medicine, Ann Arbor, MI 48109, USA; Department of Statistics, University of Michigan, Ann Arbor, MI, USA
| | - Ling Zhang
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Center for Reproductive Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jin Zhang
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University, Hangzhou, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Sriram Chandrasekaran
- Center for Bioinformatics and Computational Medicine, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Program in Chemical Biology, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
13
|
Yu P, Satyaraj E. Effect of Bovine Colostrum on Canine Immune Health. Animals (Basel) 2025; 15:185. [PMID: 39858185 PMCID: PMC11759141 DOI: 10.3390/ani15020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Colostrum, the first fluid secreted by the mammary glands of mammalian mothers, contains essential nutrients for the health and survival of newborns. Bovine colostrum (BC) is notable for its high concentrations of bioactive components, such as immunoglobulins and lactoferrin. Despite dogs being the world's most popular companion animals, there is limited research on their immune systems compared to humans. This summary aims to consolidate published studies that explore the immune benefits of BC, focusing specifically on its implications for dogs.
Collapse
Affiliation(s)
- Ping Yu
- Nestlé Purina Research, One Checkerboard Square, St. Louis, MO 63164, USA
| | - Ebenezer Satyaraj
- Nestlé Purina Research, One Checkerboard Square, St. Louis, MO 63164, USA
| |
Collapse
|
14
|
Lan T, Gao F, Cai Y, Lv Y, Zhu J, Liu H, Xie S, Wan H, He H, Xie K, Liu C, Wu H. The protein circPETH-147aa regulates metabolic reprogramming in hepatocellular carcinoma cells to remodel immunosuppressive microenvironment. Nat Commun 2025; 16:333. [PMID: 39747873 PMCID: PMC11696079 DOI: 10.1038/s41467-024-55577-0] [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: 05/27/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Metabolic reprogramming fuels cancer cell metastasis and remodels the immunosuppressive tumor microenvironment (TME). We report here that circPETH, a circular RNA (circRNA) transported via extracellular vesicles (EVs) from tumor-associated macrophages (TAMs) to hepatocellular carcinoma (HCC) cells, facilitates glycolysis and metastasis in recipient HCC cells. Mechanistically, circPETH-147aa, encoded by circPETH in an m6A-driven manner, promotes PKM2-catalyzed ALDOA-S36 phosphorylation via the MEG pocket. Furthermore, circPETH-147aa impairs anti-HCC immunity by increasing HuR-dependent SLC43A2 mRNA stability and driving methionine and leucine deficiency in cytotoxic CD8+ T cells. Importantly, through virtual and experimental screening, we find that a small molecule, Norathyriol, is an effective inhibitor that targets the MEG pocket on the circPETH-147aa surface. Norathyriol reverses circPETH-147aa-facilitated acquisition of metabolic and metastatic phenotypes by HCC cells, increases anti-PD1 efficacy, and enhances cytotoxic CD8+ T-cell function. Here we show that Norathyriol is a promising anti-HCC agent that contributes to attenuating the resistance of advanced HCC to immune checkpoint blocker (ICB) therapies.
Collapse
Affiliation(s)
- Tian Lan
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China.
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Fengwei Gao
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yunshi Cai
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yinghao Lv
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jiang Zhu
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hu Liu
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Sinan Xie
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Haifeng Wan
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Haorong He
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Kunlin Xie
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Chang Liu
- Department of Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Wu
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China.
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
15
|
Singer M, Hamdy R, Ghonaim JH, Husseiny MI. Metabolic Imbalance in Immune Cells in Relation to Metabolic Disorders, Cancer, and Infections. METABOLIC DYNAMICS IN HOST-MICROBE INTERACTION 2025:187-218. [DOI: 10.1007/978-981-96-1305-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
|
16
|
Tosello V, Rompietti C, Papathanassiu AE, Arrigoni G, Piovan E. BCAT1 Associates with DNA Repair Proteins KU70 and KU80 and Contributes to Regulate DNA Repair in T-Cell Acute Lymphoblastic Leukemia (T-ALL). Int J Mol Sci 2024; 25:13571. [PMID: 39769333 PMCID: PMC11676169 DOI: 10.3390/ijms252413571] [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] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Increased expression of branched-chain amino acid (BCAA) transaminase 1 (BCAT1) often correlates with tumor aggressiveness and drug resistance in cancer. We have recently reported that BCAT1 was overexpressed in a subgroup of T-cell acute lymphoblastic (T-ALL) samples, especially those with NOTCH1 activating mutations. Interestingly, BCAT1-depleted cells showed pronounced sensitivity to DNA-damaging agents such as etoposide; however, how BCAT1 regulates this sensitivity remains uncertain. Here, we provide further clues on its chemo-sensitizing effect. Indeed, BCAT1 protein regulates the non-homologous end joining (c-NHEJ) DNA repair pathway by physically associating with the KU70/KU80 heterodimer. BCAT1 inhibition during active repair of DNA double-strand breaks (DSBs) led to increased KU70/KU80 acetylation and impaired c-NHEJ repair, a dramatic increase in DSBs, and ultimately cell death. Our results suggest that, in T-ALL, BCAT1 possesses non-metabolic functions that confer a drug resistance mechanism and that targeting BCAT1 activity presents a novel strategy to improve chemotherapy response in T-ALL patients.
Collapse
Affiliation(s)
- Valeria Tosello
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV—IRCCS, 35127 Padua, Italy;
| | - Chiara Rompietti
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV—IRCCS, 35128 Padua, Italy;
| | | | - Giorgio Arrigoni
- Department of Biomedical Sciences, University of Padua, 35122 Padua, Italy;
- Proteomics Center, University of Padua and Azienda Ospedaliera of Padua, 35131 Padua, Italy
| | - Erich Piovan
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV—IRCCS, 35128 Padua, Italy;
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padua, Italy
| |
Collapse
|
17
|
Yu L, Bao S, Zhu F, Xu Y, Liu Y, Jiang R, Yang C, Cao F, Chen W, Li P. Association between branched-chain amino acid levels and gastric cancer risk: large-scale prospective cohort study. Front Nutr 2024; 11:1479800. [PMID: 39634548 PMCID: PMC11614650 DOI: 10.3389/fnut.2024.1479800] [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: 08/12/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024] Open
Abstract
Background Gastric cancer (GC) remains a malignancy with high incidence and mortality rates worldwide. Although branched-chain amino acids (BCAAs) play a crucial role in various physiological and pathological processes, their specific relationship with risk of GC remains unclear. Methods We conducted a large-scale prospective cohort from UK Biobank database. We evaluated the relationship between BCAA levels and risk of GC using Cox regression, Kaplan-Meier survival curves, the accelerated failure time (AFT) model, and restricted cubic spline (RCS) analysis. Results During the follow-up of 12 years, 247,753 participants were included in the study. And the Cox regression analysis revealed that higher levels of isoleucine (HR = 0.65, 95% CI 0.48-0.89; p = 0.007), leucine (HR = 0.57, 95% CI 0.42-0.79; p < 0.001), valine (HR = 0.53, 95% CI 0.39-0.73; p < 0.001), and total BCAAs were associated with a reduced risk of GC (HR = 0.51, 95% CI 0.37-0.70; p < 0.001). Kaplan-Meier curves and the AFT model confirmed that elevated BCAA levels significantly delayed the onset of GC. Additionally, RCS analysis identified nonlinear dose-response relationships between BCAAs and risk of GC. Stratified analyses indicated that the protective effect of BCAAs was consistent across various subgroups, with a more pronounced impact in older individuals without chronic diseases. Conclusion Elevated BCAA levels are significantly associated with a reduced risk of GC, particularly in older adults. This finding highlights the potential of BCAAs in GC prevention and suggests that future research and clinical practice should emphasize regulating BCAA levels.
Collapse
Affiliation(s)
- Liang Yu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei City, China
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei City, China
| | - Shiming Bao
- Department of Emergency Surgery, Tongling People's Hospital, Tongling, China
| | - Feng Zhu
- Department of General Surgery, Tongling People's Hospital, Tongling, China
| | - Yanyan Xu
- Department of Emergency Surgery, Tongling People's Hospital, Tongling, China
| | - Yanwei Liu
- Department of Emergency Surgery, Tongling People's Hospital, Tongling, China
| | - Runben Jiang
- Department of Emergency Surgery, Tongling People's Hospital, Tongling, China
| | - Chuang Yang
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Feng Cao
- Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Wei Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei City, China
| | - Pengtao Li
- Department of Emergency Medicine, No.2 People's Hospital of Fuyang City, Fuyang City, Anhui Province, China
- Department of Emergency Medicine, Fuyang Infectious Disease Clinical College of Anhui Medical University, Hefei City, China
| |
Collapse
|
18
|
Zhao M, Lin J, Wang X, Chen C, Li J, Yu J, Zhou T, Liang Y, Shen X, Shi R, Yang S, Zeng S, Deng Y, Duan X, Zhou L, Sun X, Wang Y, Shu Z. Multi-immunometabolomics mining: NP prevents hyperimmune in ALI by inhibiting Leucine/PI3K/Akt/mTOR signaling pathway. Free Radic Biol Med 2024; 225:302-315. [PMID: 39370053 DOI: 10.1016/j.freeradbiomed.2024.09.053] [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: 05/06/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Acute lung injury (ALI) is currently a global health concern. Nicandra physalodes (L.) Gaertn. (NP) holds an important position in traditional Chinese medicine and nutrition. The potential protective mechanisms of NP against ALI remain unknown. The purpose of this study was to investigate the protective effects and molecular mechanisms of NP extract (NPE) on lipopolysaccharide (LPS)-induced ALI in mice. By utilizing network pharmacology to forecast the active ingredients in NP as well as possible signaling pathways. The composition of the NPE was analyzed using UPLC-Q-TOF-MS/MS. In addition, 1H-NMR immunometabolomics was employed to identify alterations in primary metabolic pathways and metabolites in the lung, serum, and fecal tissues. Finally, the protein and gene expression of key pathways were verified by IHC, IF, RT-qPCR, and ELISA. It was found that the main ingredients of NPE were revealed to be nicandrenone, withanolide A, and baicalin. NPE significantly improved lung injury, pulmonary edema, and inflammatory cell infiltration in mice with ALI. In addition, NPE improved autophagic activity and alleviated Th1 and Th17 cell-induced lung inflammation by suppressing the PI3K/Akt/mTOR signaling pathway. Importantly, immunometabolomic analysis of fecal, serum, and lung tissues revealed that NPE reversed ALI-induced leucine resistance by remodeling immunometabolism. We confirmed NPE prevents ALI by remodeling immunometabolism, regulating the Leucine/PI3K/Akt/mTOR signaling pathway, inhibiting Th1/Th17 cell differentiation, and providing a scientific immunological basis for the clinical application of NPE.
Collapse
Affiliation(s)
- Mantong Zhao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jiazi Lin
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xiao Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Chengkai Chen
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jianhua Li
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jiamin Yu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Tong Zhou
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yefang Liang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xuejuan Shen
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ruixiang Shi
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Simin Yang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Shuting Zeng
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yongan Deng
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xiaodong Duan
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Lichang Zhou
- Ruyuan Yao Autonomous County Agricultural Technology Promotion Center, Shaoguan, 512700, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China.
| | - Yi Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Zunpeng Shu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China.
| |
Collapse
|
19
|
Akbay B, Omarova Z, Trofimov A, Sailike B, Karapina O, Molnár F, Tokay T. Double-Edge Effects of Leucine on Cancer Cells. Biomolecules 2024; 14:1401. [PMID: 39595578 PMCID: PMC11591885 DOI: 10.3390/biom14111401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Leucine is an essential amino acid that cannot be produced endogenously in the human body and therefore needs to be obtained from dietary sources. Leucine plays a pivotal role in stimulating muscle protein synthesis, along with isoleucine and valine, as the group of branched-chain amino acids, making them one of the most popular dietary supplements for athletes and gym-goers. The individual effects of leucine, however, have not been fully clarified, as most of the studies so far have focused on the grouped effects of branched-chain amino acids. In recent years, leucine and its metabolites have been shown to stimulate muscle protein synthesis mainly via the mammalian target of the rapamycin complex 1 signaling pathway, thereby improving muscle atrophy in cancer cachexia. Interestingly, cancer research suggests that leucine may have either anti-cancer or pro-tumorigenic effects. In the current manuscript, we aim to review leucine's roles in muscle protein synthesis, tumor suppression, and tumor progression, specifically summarizing the molecular mechanisms of leucine's action. The role of leucine is controversial in hepatocellular carcinoma, whereas its pro-tumorigenic effects have been demonstrated in breast and pancreatic cancers. In summary, leucine being used as nutritional supplement for athletes needs more attention, as its pro-oncogenic effects may have been identified by recent studies. Anti-cancer or pro-tumorigenic effects of leucine in various cancers should be further investigated to achieve clear conclusions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tursonjan Tokay
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Kabanbay Batyr 53, Astana 010000, Kazakhstan; (B.A.); (Z.O.); (A.T.); (B.S.); (O.K.); (F.M.)
| |
Collapse
|
20
|
Lü D, Wang Z, Wang Y, Qin S. Identification of function modules in the co-expression protein-protein interaction network of Bombyx mori in response to Beauveria bassiana infection. J Invertebr Pathol 2024; 207:108214. [PMID: 39366479 DOI: 10.1016/j.jip.2024.108214] [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: 05/27/2024] [Revised: 09/08/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Beauveria bassiana (B. bassiana) is a common fungal disease in sericulture. Previous research has primarily focused on investigating genes involved in innate immunity. However, the response of Bombyx mori (B. mori) to B. bassiana requires the coordination of other biological processes in addition to the immune system. We measured protein expression profile of B. mori after inoculating B. bassiana using iTRAQ technology in previous. Here we constructed a co-expression protein-protein interaction network of B. mori in response to B. bassiana infection. Subnetworks and modules were analyzed, and the functions of these modules were annotated. The results revealed the identification of numerous proteins associated with cellular immunity, including those involved in phagosomes, lysosomes, mTOR signaling, sugar metabolism, and the ubiquitin-proteasome pathway. Meanwhile, we observed that the pathways involved in protein synthesis were activated, including pyruvate and purine metabolism, RNA transport, ribosome, protein processing in endoplasmic reticulum, and protein export pathways, during B. bassiana infection. Based on this analysis, we selected six candidate genes (shock protein, ribosome, translocon, actin muscle-type A2, peptidoglycan recognition protein, and collagenase) that were found to be related to the response to B. bassiana. Further verification experiments demonstrated significant changes in their expression levels after inoculation with B. bassiana. These research findings provide new insights into the molecular mechanism of insect immune response to fungal infection.
Collapse
Affiliation(s)
- Dingding Lü
- Zhenjiang College, Zhenjiang 212028, China; School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China
| | - Zihe Wang
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China
| | - Ya Wang
- Zhenjiang College, Zhenjiang 212028, China
| | - Sheng Qin
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China; Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212018, China.
| |
Collapse
|
21
|
Singh D, Menghini P, Rodriguez-Palacios A, Martino LD, Cominelli F, Basson AR. Leucine-Enriched Diet Reduces Fecal MPO but Does Not Protect Against DSS Colitis in a Mouse Model of Crohn's Disease-like Ileitis. Int J Mol Sci 2024; 25:11748. [PMID: 39519299 PMCID: PMC11545852 DOI: 10.3390/ijms252111748] [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] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Understanding the complex link between inflammation, gut health, and dietary amino acids is becoming increasingly important in the pathophysiology of inflammatory bowel disease (IBD). This study tested the hypothesis that a leucine-rich diet could attenuate inflammation and improve gut health in a mouse model of IBD. Specifically, we investigated the effects of a leucine-rich diet on dextran sulfate sodium (DSS)-induced colitis in germ-free (GF) SAMP1/YitFC (SAMP) mice colonized with human gut microbiota (hGF-SAMP). hGF-SAMP mice were fed one of four different diets: standard mouse diet (CHOW), American diet (AD), leucine-rich AD (AD + AA), or leucine-rich CHOW diet (CH + AA). Body weight, myeloperoxidase (MPO) activity, gut permeability, colonoscopy scores, and histological analysis were measured. Mice on a leucine-rich CHOW diet showed a decrease in fecal MPO prior to DSS treatment as compared to those on a regular diet (p > 0.05); however, after week five, prior to DSS, this effect had diminished. Following DSS treatment, there was no significant difference in gut permeability, fecal MPO activity, or body weight changes between the leucine-supplemented and control groups. These findings suggest that while a leucine-rich diet may transiently affect fecal MPO levels in hGF-SAMP mice, it does not confer protection against DSS-induced colitis symptoms or mitigate inflammation in the long term.
Collapse
Affiliation(s)
- Drishtant Singh
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA;
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (P.M.); (A.R.-P.); (F.C.)
| | - Paola Menghini
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (P.M.); (A.R.-P.); (F.C.)
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Alexander Rodriguez-Palacios
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (P.M.); (A.R.-P.); (F.C.)
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Mouse Models Core, Silvio O’Conte Cleveland Digestive Diseases Research Core Center, Cleveland, OH 44106, USA
- Germ-Free and Gut Microbiome Core, Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Luca Di Martino
- Case Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA;
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Fabio Cominelli
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (P.M.); (A.R.-P.); (F.C.)
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Mouse Models Core, Silvio O’Conte Cleveland Digestive Diseases Research Core Center, Cleveland, OH 44106, USA
- Germ-Free and Gut Microbiome Core, Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Abigail Raffner Basson
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA;
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (P.M.); (A.R.-P.); (F.C.)
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
22
|
Hu T, Liu CH, Lei M, Zeng Q, Li L, Tang H, Zhang N. Metabolic regulation of the immune system in health and diseases: mechanisms and interventions. Signal Transduct Target Ther 2024; 9:268. [PMID: 39379377 PMCID: PMC11461632 DOI: 10.1038/s41392-024-01954-6] [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: 03/27/2024] [Revised: 07/18/2024] [Accepted: 08/11/2024] [Indexed: 10/10/2024] Open
Abstract
Metabolism, including glycolysis, oxidative phosphorylation, fatty acid oxidation, and other metabolic pathways, impacts the phenotypes and functions of immune cells. The metabolic regulation of the immune system is important in the pathogenesis and progression of numerous diseases, such as cancers, autoimmune diseases and metabolic diseases. The concept of immunometabolism was introduced over a decade ago to elucidate the intricate interplay between metabolism and immunity. The definition of immunometabolism has expanded from chronic low-grade inflammation in metabolic diseases to metabolic reprogramming of immune cells in various diseases. With immunometabolism being proposed and developed, the metabolic regulation of the immune system can be gradually summarized and becomes more and more clearer. In the context of many diseases including cancer, autoimmune diseases, metabolic diseases, and many other disease, metabolic reprogramming occurs in immune cells inducing proinflammatory or anti-inflammatory effects. The phenotypic and functional changes of immune cells caused by metabolic regulation further affect and development of diseases. Based on experimental results, targeting cellular metabolism of immune cells becomes a promising therapy. In this review, we focus on immune cells to introduce their metabolic pathways and metabolic reprogramming, and summarize how these metabolic pathways affect immune effects in the context of diseases. We thoroughly explore targets and treatments based on immunometabolism in existing studies. The challenges of translating experimental results into clinical applications in the field of immunometabolism are also summarized. We believe that a better understanding of immune regulation in health and diseases will improve the management of most diseases.
Collapse
Affiliation(s)
- Tengyue Hu
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Chang-Hai Liu
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Min Lei
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qingmin Zeng
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Li Li
- Division of Renal and endocrinology, Qin Huang Hospital, Xi'an, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China.
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Nannan Zhang
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China.
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
23
|
Yang Q, Zhu X, Huang P, Li C, Han L, Han Y, Gan R, Xin B, Tu Y, Zhou S, Yuan T, Hao J, Li C, Zhang L, Shi L, Guo C. BCKDK modification enhances the anticancer efficacy of CAR-T cells by reprogramming branched chain amino acid metabolism. Mol Ther 2024; 32:3128-3144. [PMID: 38734897 PMCID: PMC11403223 DOI: 10.1016/j.ymthe.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/25/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024] Open
Abstract
Altered branched chain amino acids (BCAAs), including leucine, isoleucine, and valine, are frequently observed in patients with advanced cancer. We evaluated the efficacy of chimeric antigen receptor (CAR) T cell-mediated cancer cell lysis potential in the immune microenvironment of BCAA supplementation and deletion. BCAA supplementation increased cancer cell killing percentage, while accelerating BCAA catabolism and decreasing BCAA transporter decreased cancer cell lysis efficacy. We thus designed BCKDK engineering CAR T cells for the reprogramming of BCAA metabolism in the tumor microenvironment based on the genotype and phenotype modification. BCKDK overexpression (OE) in CAR-T cells significantly improved cancer cell lysis, while BCKDK knockout (KO) resulted in inferior lysis potential. In an in vivo experiment, BCKDK-OE CAR-T cell treatment significantly prolonged the survival of mice bearing NALM6-GL cancer cells, with the differentiation of central memory cells and an increasing proportion of CAR-T cells in the peripheral circulation. BCKDK-KO CAR-T cell treatment resulted in shorter survival and a decreasing percentage of CAR-T cells in the peripheral circulation. In conclusion, BCKDK-engineered CAR-T cells exert a distinct phenotype for superior anticancer efficiency.
Collapse
Affiliation(s)
- Quanjun Yang
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xinting Zhu
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ping Huang
- Center for Chemical Glycobiology, Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chunyan Li
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai JiaoTong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Leng Han
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yonglong Han
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Run Gan
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Bo Xin
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yixing Tu
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Shumin Zhou
- Institution of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ting Yuan
- Department of Bone Oncology, Shanghai Sixth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Juan Hao
- Department of Endocrinology, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 230 Baoding Road, Shanghai 200082, China
| | - Chunqiong Li
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Li Zhang
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Lei Shi
- Department of Oncology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, China.
| | - Cheng Guo
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
24
|
Xiao C, Li Y, Liu Y, Dong R, He X, Lin Q, Zang X, Wang K, Xia Y, Kong L. Overcoming Cancer Persister Cells by Stabilizing the ATF4 Promoter G-quadruplex. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401748. [PMID: 38994891 PMCID: PMC11425212 DOI: 10.1002/advs.202401748] [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: 02/20/2024] [Revised: 06/23/2024] [Indexed: 07/13/2024]
Abstract
Persister cells (PS) selected for anticancer therapy have been recognized as a significant contributor to the development of treatment-resistant malignancies. It is found that imposing glutamine restriction induces the generation of PS, which paradoxically bestows heightened resistance to glutamine restriction treatment by activating the integrated stress response and initiating the general control nonderepressible 2-activating transcription factor 4-alanine, serine, cysteine-preferring transporter 2 (GCN2-ATF4-ASCT2) axis. Central to this phenomenon is the stress-induced ATF4 translational reprogramming. Unfortunately, directly targeting ATF4 protein has proven to be a formidable challenge because of its flat surface. Nonetheless, a G-quadruplex structure located within the promoter region of ATF4 (ATF4-G4) is uncovered and resolved, which functions as a transcriptional regulator and can be targeted by small molecules. The investigation identifies the natural compound coptisine (COP) as a potent binder that interacts with and stabilizes ATF4-G4. For the first time, the high-resolution structure of the COP-ATF4-G4 complex is determined. The formation of this stable complex disrupts the interaction between transcription factor AP-2 alpha (TFAP2A) and ATF4-G4, resulting in a substantial reduction in intracellular ATF4 levels and the eventual death of cancer cells. These seminal findings underscore the potential of targeting the ATF4-G4 structure to yield significant therapeutic advantages within the realm of persister cancer cells induced by glutamine-restricted therapy.
Collapse
Affiliation(s)
- Chengmei Xiao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Yipu Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Yushuang Liu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Ruifang Dong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Xiaoyu He
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Qing Lin
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Xin Zang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Kaibo Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Yuanzheng Xia
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
- Shenzhen Research Institute of China Pharmaceutical UniversityShenzhen518057China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| |
Collapse
|
25
|
Liu X, Dong M, Li Y, Li L, Zhang Y, Wang C, Wang N, Wang D. Structural properties of glucan from Russula griseocarnosa and its immunomodulatory activities mediated via T cell differentiation. Carbohydr Polym 2024; 339:122214. [PMID: 38823900 DOI: 10.1016/j.carbpol.2024.122214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 06/03/2024]
Abstract
The polysaccharide, RGP2, was isolated from Russula griseocarnosa and its immunostimulatory effects were confirmed in cyclophosphamide (CTX)-induced immunosuppressed mice. Following purification via chromatography, structural analysis revealed that RGP2 had a molecular weight of 11.82 kDa and consisted of glucose (Glc), galactose (Gal), mannose, glucuronic acid and glucosamine. Bond structure analysis and nuclear magnetic resonance characterization confirmed that the main chain of RGP2 was formed by →6)-β-D-Glcp-(1→, →3)-β-D-Glcp-(1→ and →6)-α-D-Galp-(1→, which was substituted at O-3 of →6)-β-D-Glcp-(1→ by β-D-Glcp-(1→. RGP2 was found to ameliorate pathological damage in the spleen and enhance immune cell activity in immunosuppressed mice. Based on combined multiomics analysis, RGP2 altered the abundance of immune-related microbiota (such as Lactobacillus, Faecalibacterium, and Bacteroides) in the gut and metabolites (uridine, leucine, and tryptophan) in the serum. Compared with immunosuppressed mice, RGP2 also restored the function of antigen-presenting cells, promoted the polarization of macrophages into the M1 phenotype, positively affected the differentiation of helper T cells, and inhibited regulatory T cell differentiation through the protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) pathway, ultimately exerting an immune boosting function. Overall, our findings highlight therapeutic strategies to alleviate CTX-induced immunosuppression in a clinical setting.
Collapse
Affiliation(s)
- Xin Liu
- School of Life Sciences, Jilin University, Changchun 130012, China; School of Health Science and Biomedical Engineering, Hebei University of Technology, Tianjin, 300131, China.
| | - Mingyuan Dong
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Yuan Li
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Lanzhou Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, School of Plant Protection, Jilin Agricultural University, Changchun 130118, China.
| | - Yongfeng Zhang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, School of Plant Protection, Jilin Agricultural University, Changchun 130118, China.
| | - Chunyue Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, School of Plant Protection, Jilin Agricultural University, Changchun 130118, China.
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, 6/F, 3 Sassoon Road, Pokfulam 000000, Hong Kong.
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun 130012, China; Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, School of Plant Protection, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
26
|
Liu Y, Li J, Ding C, Tong H, Yan Y, Li S, Li S, Cao Y. Leu promotes C2C12 cell differentiation by regulating the GSK3β/β-catenin signaling pathway through facilitating the interaction between SESN2 and RPN2. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:6696-6705. [PMID: 38551359 DOI: 10.1002/jsfa.13496] [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: 04/06/2023] [Revised: 03/02/2024] [Accepted: 03/29/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Leucine (Leu) is an essential amino acid that facilitates skeletal muscle satellite cell differentiation, yet its mechanism remains underexplored. Sestrin2 (SESN2) serves as a Leu sensor, binding directly to Leu, while ribophorin II (RPN2) acts as a signaling factor in multiple pathways. This study aimed to elucidate Leu's impact on mouse C2C12 cell differentiation and skeletal muscle injury repair by modulating RPN2 expression through SESN2, offering a theoretical foundation for clinical skeletal muscle injury prevention and treatment. RESULTS Leu addition promoted C2C12 cell differentiation compared to the control, enhancing early differentiation via myogenic determinant (MYOD) up-regulation. Sequencing revealed SESN2 binding to and interacting with RPN2. RPN2 overexpression up-regulated MYOD, myogenin and myosin heavy chain 2, concurrently decreased p-GSK3β and increased nuclear β-catenin. Conversely, RPN2 knockdown yielded opposite results. Combining RPN2 knockdown with Leu rescued increased p-GSK3β and decreased nuclear β-catenin compared to Leu absence. Hematoxylin and eosin staining results showed that Leu addition accelerated mouse muscle damage repair, up-regulating Pax7, MYOD and RPN2 in the cytoplasm, and nuclear β-catenin, confirming that the role of Leu in muscle injury repair was consistent with the results for C2C12 cells. CONCLUSION Leu, bound with SESN2, up-regulated RPN2 expression, activated the GSK3β/β-catenin pathway, enhanced C2C12 differentiation and expedited skeletal muscle damage repair. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yifan Liu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Jinping Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Cong Ding
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Huili Tong
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Yunqin Yan
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Shuang Li
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Shufeng Li
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| | - Yunkao Cao
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Laboratory of Cell and Development, Department of Life Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
27
|
Escolar-Peña A, Delgado-Dolset MI, Pablo-Torres C, Tarin C, Mera-Berriatua L, Cuesta Apausa MDP, González Cuervo H, Sharma R, Kho AT, Tantisira KG, McGeachie MJ, Rebollido-Rios R, Barber D, Carrillo T, Izquierdo E, Escribese MM. Specific microRNA Profile Associated with Inflammation and Lipid Metabolism for Stratifying Allergic Asthma Severity. Int J Mol Sci 2024; 25:9425. [PMID: 39273372 PMCID: PMC11394998 DOI: 10.3390/ijms25179425] [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: 07/19/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
The mechanisms underlying severe allergic asthma are complex and unknown, meaning it is a challenge to provide the most appropriate treatment. This study aimed to identify novel biomarkers for stratifying allergic asthmatic patients according to severity, and to uncover the biological mechanisms that lead to the development of the severe uncontrolled phenotype. By using miRNA PCR panels, we analyzed the expression of 752 miRNAs in serum samples from control subjects (n = 15) and mild (n = 11) and severe uncontrolled (n = 10) allergic asthmatic patients. We identified 40 differentially expressed miRNAs between severe uncontrolled and mild allergic asthmatic patients. Functional enrichment analysis revealed signatures related to inflammation, angiogenesis, lipid metabolism and mRNA regulation. A random forest classifier trained with DE miRNAs achieved a high accuracy of 97% for severe uncontrolled patient stratification. Validation of the identified biomarkers was performed on a subset of allergic asthmatic patients from the CAMP cohort at Brigham and Women's Hospital, Harvard Medical School. Four of these miRNAs (hsa-miR-99b-5p, hsa-miR-451a, hsa-miR-326 and hsa-miR-505-3p) were validated, pointing towards their potential as biomarkers for stratifying allergic asthmatic patients by severity and providing insights into severe uncontrolled asthma molecular pathways.
Collapse
Affiliation(s)
- Andrea Escolar-Peña
- Department of Basic Medical Sciences, Institute for Applied Molecular Medicine Nemesio Díez, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | - María Isabel Delgado-Dolset
- Department of Basic Medical Sciences, Institute for Applied Molecular Medicine Nemesio Díez, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | - Carmela Pablo-Torres
- Department of Basic Medical Sciences, Institute for Applied Molecular Medicine Nemesio Díez, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | - Carlos Tarin
- R+D Department, Atrys Health, 08025 Madrid, Spain
| | - Leticia Mera-Berriatua
- Department of Basic Medical Sciences, Institute for Applied Molecular Medicine Nemesio Díez, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | | | - Heleia González Cuervo
- Allergy Service, Hospital Universitario de Gran Canaria Doctor Negrín, 35010 Las Palmas de Gran Canaria, Spain
| | - Rinku Sharma
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alvin T Kho
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Kelan G Tantisira
- Division of Pediatric Respiratory Medicine, University of California San Diego and Rady Children's Hospital, San Diego, CA 92123, USA
| | - Michael J McGeachie
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rocio Rebollido-Rios
- Department I of Internal Medicine, Centre of Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50923 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50923 Cologne, Germany
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50923 Cologne, Germany
| | - Domingo Barber
- Department of Basic Medical Sciences, Institute for Applied Molecular Medicine Nemesio Díez, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | - Teresa Carrillo
- Allergy Service, Hospital Universitario de Gran Canaria Doctor Negrín, 35010 Las Palmas de Gran Canaria, Spain
| | - Elena Izquierdo
- Department of Basic Medical Sciences, Institute for Applied Molecular Medicine Nemesio Díez, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | - María M Escribese
- Department of Basic Medical Sciences, Institute for Applied Molecular Medicine Nemesio Díez, School of Medicine, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| |
Collapse
|
28
|
Yamamoto W, Hamada T, Suzuki J, Matsuoka Y, Omori-Miyake M, Kuwahara M, Matsumoto A, Nomura S, Konishi A, Yorozuya T, Yamashita M. Suppressive effect of the anesthetic propofol on the T cell function and T cell-dependent immune responses. Sci Rep 2024; 14:19337. [PMID: 39164311 PMCID: PMC11336218 DOI: 10.1038/s41598-024-69987-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024] Open
Abstract
General anesthesia is thought to suppress the immune system and negatively affect postoperative infection and the long-term prognosis of cancer. However, the mechanism underlying immunosuppression induced by general anesthetics remains unclear. In this study, we focused on propofol, which is widely used for sedation under general anesthesia and intensive care and examined its effects on the T cell function and T cell-dependent immune responses. We found that propofol suppressed T cell glycolytic metabolism, differentiation into effector T cells, and cytokine production by effector T cells. CD8 T cells activated and differentiated into effector cells in the presence of propofol in vitro showed reduced antitumor activity. Furthermore, propofol treatment suppressed the increase in the number of antigen-specific CD8 T cells during Listeria infection. In contrast, the administration of propofol improved inflammatory conditions in mouse models of inflammatory diseases, such as OVA-induced allergic airway inflammation, hapten-induced contact dermatitis, and experimental allergic encephalomyelitis. These results suggest that propofol may reduce tumor and infectious immunity by suppressing the T cell function and T cell-dependent immune responses while improving the pathogenesis and prognosis of chronic inflammatory diseases by suppressing inflammation.
Collapse
Affiliation(s)
- Waichi Yamamoto
- Department of Anesthesia and Perioperative Medicine, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
- Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Taisuke Hamada
- Department of Anesthesia and Perioperative Medicine, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
- Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Junpei Suzuki
- Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Yuko Matsuoka
- Translational Research Center, Ehime University Hospital, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Miyuki Omori-Miyake
- Department of Infections and Host Defenses, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Makoto Kuwahara
- Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Akira Matsumoto
- Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Shunsuke Nomura
- Department of Immuno-Drug Chemistry, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Amane Konishi
- Department of Anesthesia and Perioperative Medicine, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Toshihiro Yorozuya
- Department of Anesthesia and Perioperative Medicine, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Masakatsu Yamashita
- Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan.
- Translational Research Center, Ehime University Hospital, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan.
- Department of Infections and Host Defenses, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan.
- Department of Immuno-Drug Chemistry, Graduate School of Medicine, Ehime University, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan.
| |
Collapse
|
29
|
Kang YJ, Song W, Lee SJ, Choi SA, Chae S, Yoon BR, Kim HY, Lee JH, Kim C, Cho JY, Kim HJ, Lee WW. Inhibition of BCAT1-mediated cytosolic leucine metabolism regulates Th17 responses via the mTORC1-HIF1α pathway. Exp Mol Med 2024; 56:1776-1790. [PMID: 39085353 PMCID: PMC11372109 DOI: 10.1038/s12276-024-01286-z] [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: 11/30/2023] [Revised: 03/26/2024] [Accepted: 04/19/2024] [Indexed: 08/02/2024] Open
Abstract
Branched-chain amino acids (BCAAs), particularly leucine, are indispensable AAs for immune regulation through metabolic rewiring. However, the molecular mechanism underlying this phenomenon remains unclear. Our investigation revealed that T-cell receptor (TCR)-activated human CD4+ T cells increase the expression of BCAT1, a cytosolic enzyme responsible for BCAA catabolism, and SLC7A5, a major BCAA transporter. This upregulation facilitates increased leucine influx and catabolism, which are particularly crucial for Th17 responses. Activated CD4+ T cells induce an alternative pathway of cytosolic leucine catabolism, generating a pivotal metabolite, β-hydroxy β-methylbutyric acid (HMB), by acting on BCAT1 and 4-hydroxyphenylpyruvate dioxygenase (HPD)/HPD-like protein (HPDL). Inhibition of BCAT1-mediated cytosolic leucine metabolism, either with BCAT1 inhibitor 2 (Bi2) or through BCAT1, HPD, or HPDL silencing using shRNA, attenuates IL-17 production, whereas HMB supplementation abrogates this effect. Mechanistically, HMB contributes to the regulation of the mTORC1-HIF1α pathway, a major signaling pathway for IL-17 production, by increasing the mRNA expression of HIF1α. This finding was corroborated by the observation that treatment with L-β-homoleucine (LβhL), a leucine analog and competitive inhibitor of BCAT1, decreased IL-17 production by TCR-activated CD4+ T cells. In an in vivo experimental autoimmune encephalomyelitis (EAE) model, blockade of BCAT1-mediated leucine catabolism, either through a BCAT1 inhibitor or LβhL treatment, mitigated EAE severity by decreasing HIF1α expression and IL-17 production in spinal cord mononuclear cells. Our findings elucidate the role of BCAT1-mediated cytoplasmic leucine catabolism in modulating IL-17 production via HMB-mediated regulation of mTORC1-HIF1α, providing insights into its relevance to inflammatory conditions.
Collapse
Affiliation(s)
- Yeon Jun Kang
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Woorim Song
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Su Jeong Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Seung Ah Choi
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Sihyun Chae
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University, College of Medicine and Hospital, Seoul, 03080, Republic of Korea
| | - Bo Ruem Yoon
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Hee Young Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jung Ho Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080, Korea
| | - Chulwoo Kim
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Joo-Youn Cho
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University, College of Medicine and Hospital, Seoul, 03080, Republic of Korea
| | - Hyun Je Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, 03080, Korea
| | - Won-Woo Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Seoul National University Cancer Research Institute, Institue of Endemic Diseases and Ischemic/Hypoxic Disease Institute, Seoul National University Medical Research Center, Seoul National University Hospital Biomedical Research Institute, Seoul, 03080, Republic of Korea.
| |
Collapse
|
30
|
Jana SK, Som NN, Jha PK. Size-Dependent Fullerenes for Enhanced Interaction of l-Leucine: A Combined DFT and MD Simulations Approach. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:13844-13859. [PMID: 38916256 DOI: 10.1021/acs.langmuir.4c00806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Fullerene-based biosensors have received great attention due to their unique electronic properties that allow them to transduce electrical signals by accepting electrons from amino acids. Babies with MSUD (maple syrup urine disease) are unable to break down amino acids such as l-leucine, and excess levels of the l-leucine are harmful. Therefore, sensing of l-leucine is foremost required. We aim to investigate the interaction tendencies of size-variable fullerenes (CX; X = 24, 36, 50, and 70) toward l-leucine (LEU) using density functional theory (DFT-D3) and classical molecular dynamics (MD) simulation. The C24 fullerene shows the highest affinity of the LEU biomolecule in the gas phase. Smaller fullerenes (C24 and C36) show stronger interactions with leucine due to their higher curvature in water environments. Moreover, recovery times in the ranges of 1010 and 104 s make it a viable candidate for the isolation application of LEU from the biological system. Further, the interaction between LEU and fullerenes is in line with the natural bond order (NBO) analysis, Mulliken charge analysis, quantum theory atom in molecule (QTAIM) analysis, and reduced density gradient (RDG) analysis. At 310 K, employing the explicit water model in classical MD simulations, fullerenes C24 and C36 demonstrate notably elevated binding free energies (-24.946 kJ/mol) in relation to LEU, showcasing their potential as sensors for l-leucine. Here, we demonstrate that the smaller fullerene exhibits a higher potential for l-leucine sensors than the larger fullerene.
Collapse
Affiliation(s)
- Sourav Kanti Jana
- Department of Physics, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 39002, India
| | - Narayan N Som
- Institute of High-Pressure Physics, Polish Academy of Sciences, Sokolowska 29/37, 01-142 Warsaw, Poland
| | - Prafulla K Jha
- Department of Physics, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 39002, India
| |
Collapse
|
31
|
Gusdon AM, Savarraj JPJ, Feng D, Starkman A, Li G, Bodanapally U, Zimmerman W, Ryan AS, Choi HA, Badjatia N. Identification of metabolites associated with preserved muscle volume after aneurysmal subarachnoid hemorrhage due to high protein supplementation and neuromuscular electrical stimulation. Sci Rep 2024; 14:15071. [PMID: 38956192 PMCID: PMC11219968 DOI: 10.1038/s41598-024-64666-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/11/2024] [Indexed: 07/04/2024] Open
Abstract
The INSPIRE randomized clinical trial demonstrated that a high protein diet (HPRO) combined with neuromuscular electrical stimulation (NMES) attenuates muscle atrophy and may improve outcomes after aneurysmal subarachnoid hemorrhage We sought to identify specific metabolites mediating these effects. Blood samples were collected from subjects on admission prior to randomization to either standard of care (SOC; N = 12) or HPRO + NMES (N = 12) and at 7 days. Untargeted metabolomics were performed for each plasma sample. Sparse partial least squared discriminant analysis identified metabolites differentiating each group. Correlation coefficients were calculated between each metabolite and total protein per day and muscle volume. Multivariable models determined associations between metabolites and muscle volume. Unique metabolites (18) were identified differentiating SOC from HPRO + NMES. Of these, 9 had significant positive correlations with protein intake. In multivariable models, N-acetylleucine was significantly associated with preserved temporalis [OR 1.08 (95% CI 1.01, 1.16)] and quadricep [OR 1.08 (95% CI 1.02, 1.15)] muscle volume. Quinolinate was also significantly associated with preserved temporalis [OR 1.05 (95% CI 1.01, 1.09)] and quadricep [OR 1.04 (95% CI 1.00, 1.07)] muscle volume. N-acetylserine and β-hydroxyisovaleroylcarnitine were associated with preserved temporalis or quadricep volume. Metabolites defining HPRO + NMES had strong correlations with protein intake and were associated with preserved muscle volume.
Collapse
Affiliation(s)
- Aaron M Gusdon
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Jude P J Savarraj
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Diana Feng
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Adam Starkman
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Guoyan Li
- Division of Gerontology, Geriatric, and Palliative Medicine, Department of Medicine, Geriatric Research, Education, and Clinical Center (GRECC), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Uttam Bodanapally
- Department of Radiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - William Zimmerman
- Program in Trauma, Shock Trauma Neurocritical Care and Department of Neurology, University of Maryland School of Medicine, 22 S. Greene Street, G7K19, Baltimore, MD, 21201, USA
| | - Alice S Ryan
- Division of Gerontology, Geriatric, and Palliative Medicine, Department of Medicine, Geriatric Research, Education, and Clinical Center (GRECC), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Huimahn A Choi
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Neeraj Badjatia
- Program in Trauma, Shock Trauma Neurocritical Care and Department of Neurology, University of Maryland School of Medicine, 22 S. Greene Street, G7K19, Baltimore, MD, 21201, USA.
| |
Collapse
|
32
|
Veliz K, Shen F, Shestova O, Shestov M, Shestov A, Sleiman S, Hansen T, O’Connor RS, Gill S. Deletion of CD38 enhances CD19 chimeric antigen receptor T cell function. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200819. [PMID: 38912091 PMCID: PMC11193011 DOI: 10.1016/j.omton.2024.200819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/22/2024] [Indexed: 06/25/2024]
Abstract
Cell surface molecules transiently upregulated on activated T cells can play a counter-regulatory role by inhibiting T cell function. Deletion or blockade of such immune checkpoint receptors has been investigated to improve the function of engineered immune effector cells. CD38 is upregulated on activated T cells, and although there have been studies showing that CD38 can play an inhibitory role in T cells, how it does so has not fully been elucidated. In comparison with molecules such as PD1, CTLA4, LAG3, and TIM3, we found that CD38 displays more sustained and intense expression following acute activation. After deleting CD38 from human chimeric antigen receptor (CAR) T cells, we showed relative resistance to exhaustion in vitro and improved anti-tumor function in vivo. CD38 is a multifunctional ectoenzyme with hydrolase and cyclase activities. Reintroduction of CD38 mutants into T cells lacking CD38 provided further evidence supporting the understanding that CD38 plays a crucial role in producing the immunosuppressive metabolite adenosine and utilizing nicotinamide adenine dinucleotide (NAD) in human T cells. Taken together, these results highlight a role for CD38 as an immunometabolic checkpoint in T cells and lead us to propose CD38 deletion as an additional avenue for boosting CAR T cell function.
Collapse
Affiliation(s)
- Kimberly Veliz
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Feng Shen
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Olga Shestova
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maksim Shestov
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexander Shestov
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sara Sleiman
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tyler Hansen
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roddy S. O’Connor
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Saar Gill
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cell Therapy and Transplant Program, Division of Hematology-Oncology and Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
33
|
Gavade A, Nagraj AK, Patel R, Pais R, Dhanure P, Scheele J, Seiz W, Patil J. Understanding the Specific Implications of Amino Acids in the Antibody Development. Protein J 2024; 43:405-424. [PMID: 38724751 DOI: 10.1007/s10930-024-10201-4] [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] [Accepted: 04/21/2024] [Indexed: 06/01/2024]
Abstract
As the demand for immunotherapy to treat and manage cancers, infectious diseases and other disorders grows, a comprehensive understanding of amino acids and their intricate role in antibody engineering has become a prime requirement. Naturally produced antibodies may not have the most suitable amino acids at the complementarity determining regions (CDR) and framework regions, for therapeutic purposes. Therefore, to enhance the binding affinity and therapeutic properties of an antibody, the specific impact of certain amino acids on the antibody's architecture must be thoroughly studied. In antibody engineering, it is crucial to identify the key amino acid residues that significantly contribute to improving antibody properties. Therapeutic antibodies with higher binding affinity and improved functionality can be achieved through modifications or substitutions with highly suitable amino acid residues. Here, we have indicated the frequency of amino acids and their association with the binding free energy in CDRs. The review also analyzes the experimental outcome of two studies that reveal the frequency of amino acids in CDRs and provides their significant correlation between the outcomes. Additionally, it discusses the various bond interactions within the antibody structure and antigen binding. A detailed understanding of these amino acid properties should assist in the analysis of antibody sequences and structures needed for designing and enhancing the overall performance of therapeutic antibodies.
Collapse
Affiliation(s)
- Akshata Gavade
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Anil Kumar Nagraj
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Riya Patel
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Roylan Pais
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Pratiksha Dhanure
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | | | | | - Jaspal Patil
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India.
| |
Collapse
|
34
|
Braden ML, Gwin JA, Leidy HJ. Examining the Direct and Indirect Effects of Postprandial Amino Acid Responses on Markers of Satiety following the Acute Consumption of Lean Beef-Rich Meals in Healthy Women with Overweight. Nutrients 2024; 16:1718. [PMID: 38892651 PMCID: PMC11174850 DOI: 10.3390/nu16111718] [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: 04/30/2024] [Revised: 05/25/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
The consumption of protein-rich foods stimulates satiety more than other macronutrient-rich foods; however, the underlying mechanisms-of-action are not well-characterized. The objective of this study was to identify the direct and indirect effects of postprandial amino acid (AA) responses on satiety. Seventeen women (mean ± SEM, age: 33 ± 1 year; BMI: 27.8 ± 0.1 kg/m2) consumed a eucaloric, plant-based diet containing two servings of lean beef/day (i.e., 7.5 oz (207 g)) for 7 days. During day 6, the participants completed a 12 h controlled-feeding, clinical testing day including repeated satiety questionnaires and blood sampling to assess pre- and postprandial plasma AAs, PYY, and GLP-1. Regression and mediation analyses were completed to assess AA predictors and hormonal mediators. Total plasma AAs explained 41.1% of the variance in perceived daily fullness (p < 0.001), 61.0% in PYY (p < 0.001), and 66.1% in GLP-1 (p < 0.001) concentrations, respectively. Several individual AAs significantly predicted fluctuations in daily fullness, PYY, and GLP-1. In completing mediation analyses, the effect of plasma leucine on daily fullness was fully mediated by circulating PYY concentrations (indirect effect = B: 0.09 [Boot 95% CI: 0.032, 0.17]) as no leucine-fullness direct effect was observed. No other mediators were identified. Although a number of circulating AAs predict satiety, leucine was found to do so through changes in PYY concentrations in middle-aged women.
Collapse
Affiliation(s)
- Morgan L. Braden
- Department of Nutritional Sciences, University of Texas at Austin, Austin, TX 78723, USA;
- Department of Pediatrics, Dell Medical School, University of Texas at Austin, Austin, TX 78723, USA
| | - Jess A. Gwin
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA 01760, USA;
| | - Heather J. Leidy
- Department of Nutritional Sciences, University of Texas at Austin, Austin, TX 78723, USA;
- Department of Pediatrics, Dell Medical School, University of Texas at Austin, Austin, TX 78723, USA
| |
Collapse
|
35
|
Wiebe M, Milligan K, Brewer J, Fuentes AM, Ali-Adeeb R, Brolo AG, Lum JJ, Andrews JL, Haston C, Jirasek A. Metabolic profiling of murine radiation-induced lung injury with Raman spectroscopy and comparative machine learning. Analyst 2024; 149:2864-2876. [PMID: 38619825 DOI: 10.1039/d4an00152d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Radiation-induced lung injury (RILI) is a dose-limiting toxicity for cancer patients receiving thoracic radiotherapy. As such, it is important to characterize metabolic associations with the early and late stages of RILI, namely pneumonitis and pulmonary fibrosis. Recently, Raman spectroscopy has shown utility for the differentiation of pneumonitic and fibrotic tissue states in a mouse model; however, the specific metabolite-disease associations remain relatively unexplored from a Raman perspective. This work harnesses Raman spectroscopy and supervised machine learning to investigate metabolic associations with radiation pneumonitis and pulmonary fibrosis in a mouse model. To this end, Raman spectra were collected from lung tissues of irradiated/non-irradiated C3H/HeJ and C57BL/6J mice and labelled as normal, pneumonitis, or fibrosis, based on histological assessment. Spectra were decomposed into metabolic scores via group and basis restricted non-negative matrix factorization, classified with random forest (GBR-NMF-RF), and metabolites predictive of RILI were identified. To provide comparative context, spectra were decomposed and classified via principal component analysis with random forest (PCA-RF), and full spectra were classified with a convolutional neural network (CNN), as well as logistic regression (LR). Through leave-one-mouse-out cross-validation, we observed that GBR-NMF-RF was comparable to other methods by measure of accuracy and log-loss (p > 0.10 by Mann-Whitney U test), and no methodology was dominant across all classification tasks by measure of area under the receiver operating characteristic curve. Moreover, GBR-NMF-RF results were directly interpretable and identified collagen and specific collagen precursors as top fibrosis predictors, while metabolites with immune and inflammatory functions, such as serine and histidine, were top pneumonitis predictors. Further support for GBR-NMF-RF and the identified metabolite associations with RILI was found as CNN interpretation heatmaps revealed spectral regions consistent with these metabolites.
Collapse
Affiliation(s)
- Mitchell Wiebe
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| | - Kirsty Milligan
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| | - Joan Brewer
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| | - Alejandra M Fuentes
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| | - Ramie Ali-Adeeb
- Department of Chemistry, The University of Victoria, Victoria, Canada
| | - Alexandre G Brolo
- Department of Chemistry, The University of Victoria, Victoria, Canada
| | - Julian J Lum
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, Canada
| | - Jeffrey L Andrews
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| | - Christina Haston
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| | - Andrew Jirasek
- Department of Computer Science, Mathematics, Physics, and Statistics, The University of British Columbia Okanagan Campus, Kelowna, Canada.
| |
Collapse
|
36
|
Noble J, Macek Jilkova Z, Aspord C, Malvezzi P, Fribourg M, Riella LV, Cravedi P. Harnessing Immune Cell Metabolism to Modulate Alloresponse in Transplantation. Transpl Int 2024; 37:12330. [PMID: 38567143 PMCID: PMC10985621 DOI: 10.3389/ti.2024.12330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 03/06/2024] [Indexed: 04/04/2024]
Abstract
Immune cell metabolism plays a pivotal role in shaping and modulating immune responses. The metabolic state of immune cells influences their development, activation, differentiation, and overall function, impacting both innate and adaptive immunity. While glycolysis is crucial for activation and effector function of CD8 T cells, regulatory T cells mainly use oxidative phosphorylation and fatty acid oxidation, highlighting how different metabolic programs shape immune cells. Modification of cell metabolism may provide new therapeutic approaches to prevent rejection and avoid immunosuppressive toxicities. In particular, the distinct metabolic patterns of effector and suppressive cell subsets offer promising opportunities to target metabolic pathways that influence immune responses and graft outcomes. Herein, we review the main metabolic pathways used by immune cells, the techniques available to assay immune metabolism, and evidence supporting the possibility of shifting the immune response towards a tolerogenic profile by modifying energetic metabolism.
Collapse
Affiliation(s)
- Johan Noble
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, University Hospital Grenoble, Grenoble, France
- Inserm U 1209, CNRS UMR 5309, Team Epigenetics, Immunity, Metabolism, Cell Signaling and Cancer, Institute for Advanced Biosciences Grenoble, University Grenoble Alpes, La Tronche, France
| | - Zuzana Macek Jilkova
- Inserm U 1209, CNRS UMR 5309, Team Epigenetics, Immunity, Metabolism, Cell Signaling and Cancer, Institute for Advanced Biosciences Grenoble, University Grenoble Alpes, La Tronche, France
- Hepato-Gastroenterology and Digestive Oncology Department, University Hospital Grenoble, Grenoble, France
| | - Caroline Aspord
- Inserm U 1209, CNRS UMR 5309, Team Epigenetics, Immunity, Metabolism, Cell Signaling and Cancer, Institute for Advanced Biosciences Grenoble, University Grenoble Alpes, La Tronche, France
- Établissement Français du Sang Auvergne-Rhône-Alpes, R&D-Laboratory, Grenoble, France
| | - Paolo Malvezzi
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, University Hospital Grenoble, Grenoble, France
| | - Miguel Fribourg
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai New York, New York, NY, United States
| | - Leonardo V. Riella
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Paolo Cravedi
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai New York, New York, NY, United States
| |
Collapse
|
37
|
Yan H, Liu Y, Li X, Yu B, He J, Mao X, Yu J, Huang Z, Luo Y, Luo J, Wu A, Chen D. Leucine alleviates cytokine storm syndrome by regulating macrophage polarization via the mTORC1/LXRα signaling pathway. eLife 2024; 12:RP89750. [PMID: 38442142 PMCID: PMC10942637 DOI: 10.7554/elife.89750] [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] [Indexed: 03/07/2024] Open
Abstract
Cytokine storms are associated with severe pathological damage and death in some diseases. Excessive activation of M1 macrophages and the subsequent secretion of pro-inflammatory cytokines are a major cause of cytokine storms. Therefore, promoting the polarization of M2 macrophages to restore immune balance is a promising therapeutic strategy for treating cytokine storm syndrome (CSS). This study was aimed at investigating the potential protective effects of leucine on lipopolysaccharide (LPS)-induced CSS in mice and exploring the underlying mechanisms. CSS was induced by LPS administration in mice, which were concurrently administered leucine orally. In vitro, bone marrow derived macrophages (BMDMs) were polarized to M1 and M2 phenotypes with LPS and interleukin-4 (IL-4), respectively, and treated with leucine. Leucine decreased mortality in mice treated with lethal doses of LPS. Specifically, leucine decreased M1 polarization and promoted M2 polarization, thus diminishing pro-inflammatory cytokine levels and ameliorating CSS in mice. Further studies revealed that leucine-induced macrophage polarization through the mechanistic target of rapamycin complex 1 (mTORC1)/liver X receptor α (LXRα) pathway, which synergistically enhanced the expression of the IL-4-induced M2 marker Arg1 and subsequent M2 polarization. In summary, this study revealed that leucine ameliorates CSS in LPS mice by promoting M2 polarization through the mTORC1/LXRα/Arg1 signaling pathway. Our findings indicate that a fundamental link between metabolism and immunity contributes to the resolution of inflammation and the repair of damaged tissues.
Collapse
Affiliation(s)
- Hui Yan
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Yao Liu
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Xipeng Li
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Bing Yu
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Jun He
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Xiangbing Mao
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Jie Yu
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Zhiqing Huang
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Yuheng Luo
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Junqiu Luo
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Aimin Wu
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Daiwen Chen
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| |
Collapse
|
38
|
Liu S, Mochizuki M, Suzuki Y, Takemasa E, Yano A, Imai M, Mogi M. Dietary leucine supplementation restores T-cell mitochondrial respiration and regulates T-lineage differentiation in denervation-induced sarcopenic mice. J Nutr Biochem 2024; 124:109508. [PMID: 37898392 DOI: 10.1016/j.jnutbio.2023.109508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
With the aim of offsetting immune dysfunction preceded by sarcopenia, the feasibility and efficiency of nutritional leucine supplementation were evaluated using a murine denervation-induced sarcopenia model. Sciatic nerve axotomy caused significant loss of skeletal muscle of the hind limbs and accelerated mitochondrial stress along with suppressed ATP production in spleen-derived T cells. Dietary leucine intake not only ameliorated muscle mass anabolism in a sarcopenic state, but also restored mitochondrial respiratory function, as indicated by elevated levels of basal respiration, maximal respiration, spare respiratory capacity, and ATP production, in T cells, which in turn led to downregulated expression of mTOR and downstream signals, as indicated by the findings of comprehensive transcriptome analysis. Consequentially, this finally resulted in amelioration of the sarcopenia-induced relative Th1/Th17-dominant proinflammatory microenvironment. These results highlight the importance of leucine-promoted metabolic cues in directing T cell fate in a sarcopenic microenvironment. The present study provides insights that particularly help rationalize the design and optimization of leucine supplementation for chronic sarcopenic patients with autoimmune diseases.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Pharmacology, Ehime University Graduate School of Medicine, Shitsugawa, Toon, Ehime, Japan.
| | - Marii Mochizuki
- Department of Pharmacology, Ehime University Graduate School of Medicine, Shitsugawa, Toon, Ehime, Japan
| | - Yasuyuki Suzuki
- Department of Pharmacology, Ehime University Graduate School of Medicine, Shitsugawa, Toon, Ehime, Japan; Department of Anesthesiology, Saiseikai Matsuyama Hospital, Matsuyama, Japan; Research Division, Saiseikai Research Institute of Health Care and Welfare, Tokyo, Japan
| | - Erika Takemasa
- Department of Pharmacology, Ehime University Graduate School of Medicine, Shitsugawa, Toon, Ehime, Japan
| | - Akiko Yano
- Department of Pharmacology, Ehime University Graduate School of Medicine, Shitsugawa, Toon, Ehime, Japan
| | - Matome Imai
- Department of Pharmacology, Ehime University Graduate School of Medicine, Shitsugawa, Toon, Ehime, Japan
| | - Masaki Mogi
- Department of Pharmacology, Ehime University Graduate School of Medicine, Shitsugawa, Toon, Ehime, Japan
| |
Collapse
|
39
|
Oyarbide U, Crane GM, Corey SJ. The metabolic basis of inherited neutropenias. Br J Haematol 2024; 204:45-55. [PMID: 38049194 DOI: 10.1111/bjh.19192] [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: 08/29/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 12/06/2023]
Abstract
Neutrophils are the shortest-lived blood cells, which requires a prodigious degree of proliferation and differentiation to sustain physiologically sufficient numbers and be poised to respond quickly to infectious emergencies. More than 107 neutrophils are produced every minute in an adult bone marrow-a process that is tightly regulated by a small group of cytokines and chemical mediators and dependent on nutrients and energy. Like granulocyte colony-stimulating factor, the primary growth factor for granulopoiesis, they stimulate signalling pathways, some affecting metabolism. Nutrient or energy deficiency stresses the survival, proliferation, and differentiation of neutrophils and their precursors. Thus, it is not surprising that monogenic disorders related to metabolism exist that result in neutropenia. Among these are pathogenic mutations in HAX1, G6PC3, SLC37A4, TAFAZZIN, SBDS, EFL1 and the mitochondrial disorders. These mutations perturb carbohydrate, lipid and/or protein metabolism. We hypothesize that metabolic disturbances may drive the pathogenesis of a subset of inherited neutropenias just as defects in DNA damage response do in Fanconi anaemia, telomere maintenance in dyskeratosis congenita and ribosome formation in Diamond-Blackfan anaemia. Greater understanding of metabolic pathways in granulopoiesis will identify points of vulnerability in production and may point to new strategies for the treatment of neutropenias.
Collapse
Affiliation(s)
- Usua Oyarbide
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Pediatrics, Cleveland Clinic, Cleveland, Ohio, USA
| | - Genevieve M Crane
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Seth J Corey
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Pediatrics, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
40
|
Zhao L, Ma D, Wang L, Su X, Feng L, Zhu L, Chen Y, Hao Y, Wang X, Feng J. Metabolic changes with the occurrence of atherosclerotic plaques and the effects of statins. Front Immunol 2023; 14:1301051. [PMID: 38143759 PMCID: PMC10739339 DOI: 10.3389/fimmu.2023.1301051] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 11/23/2023] [Indexed: 12/26/2023] Open
Abstract
Atherosclerosis is a common cardiovascular disease caused by the abnormal expression of multiple factors and genes influenced by both environmental and genetic factors. The primary manifestation of atherosclerosis is plaque formation, which occurs when inflammatory cells consume excess lipids, affecting their retention and modification within the arterial intima. This triggers endothelial cell (EC) activation, immune cell infiltration, vascular smooth muscle cell (VSMC) proliferation and migration, foam cell formation, lipid streaks, and fibrous plaque development. These processes can lead to vascular wall sclerosis, lumen stenosis, and thrombosis. Immune cells, ECs, and VSMCs in atherosclerotic plaques undergo significant metabolic changes and inflammatory responses. The interaction of cytokines and chemokines secreted by these cells leads to the onset, progression, and regression of atherosclerosis. The regulation of cell- or cytokine-based immune responses is a novel therapeutic approach for atherosclerosis. Statins are currently the primary pharmacological agents utilised for managing unstable plaques owing to their ability to enhance endothelial function, regulate VSMC proliferation and apoptosis by reducing cholesterol levels, and mitigate the expression and activity of inflammatory cytokines. In this review, we provide an overview of the metabolic changes associated with atherosclerosis, describe the effects of inflammatory responses on atherosclerotic plaques, and discuss the mechanisms through which statins contribute to plaque stabilisation. Additionally, we examine the role of statins in combination with other drugs in the management of atherosclerosis.
Collapse
Affiliation(s)
| | - Di Ma
- Bethune First Hospital, Jilin University, Changchun, China
| | - LiJuan Wang
- Bethune First Hospital, Jilin University, Changchun, China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Khalil MI, Ali MM, Holail J, Houssein M. Growth or death? Control of cell destiny by mTOR and autophagy pathways. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 185:39-55. [PMID: 37944568 DOI: 10.1016/j.pbiomolbio.2023.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/08/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
One of the central regulators of cell growth, proliferation, and metabolism is the mammalian target of rapamycin, mTOR, which exists in two structurally and functionally different complexes: mTORC1 and mTORC2; unlike m TORC2, mTORC1 is activated in response to the sufficiency of nutrients and is inhibited by rapamycin. mTOR complexes have critical roles not only in protein synthesis, gene transcription regulation, proliferation, tumor metabolism, but also in the regulation of the programmed cell death mechanisms such as autophagy and apoptosis. Autophagy is a conserved catabolic mechanism in which damaged molecules are recycled in response to nutrient starvation. Emerging evidence indicates that the mTOR signaling pathway is frequently activated in tumors. In addition, dysregulation of autophagy was associated with the development of a variety of human diseases, such as cancer and aging. Since mTOR can inhibit the induction of the autophagic process from the early stages of autophagosome formation to the late stage of lysosome degradation, the use of mTOR inhibitors to regulate autophagy could be considered a potential therapeutic option. The present review sheds light on the mTOR and autophagy signaling pathways and the mechanisms of regulation of mTOR-autophagy.
Collapse
Affiliation(s)
- Mahmoud I Khalil
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut, 11072809, Lebanon; Molecular Biology Unit, Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt.
| | - Mohamad M Ali
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23, Uppsala, Sweden.
| | - Jasmine Holail
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia; Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom.
| | - Marwa Houssein
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut, 11072809, Lebanon.
| |
Collapse
|
42
|
Shen X, Niu N, Xue J. Oncogenic KRAS triggers metabolic reprogramming in pancreatic ductal adenocarcinoma. J Transl Int Med 2023; 11:322-329. [PMID: 38130635 PMCID: PMC10732496 DOI: 10.2478/jtim-2022-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with an extremely high lethality rate. Oncogenic KRAS activation has been proven to be a key driver of PDAC initiation and progression. There is increasing evidence that PDAC cells undergo extensive metabolic reprogramming to adapt to their extreme energy and biomass demands. Cell-intrinsic factors, such as KRAS mutations, are able to trigger metabolic rewriting. Here, we update recent advances in KRAS-driven metabolic reprogramming and the associated metabolic therapeutic potential in PDAC.
Collapse
Affiliation(s)
- Xuqing Shen
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai200127, China
| | - Ningning Niu
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai200127, China
| | - Jing Xue
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai200127, China
| |
Collapse
|
43
|
Rehman SU, Ali R, Zhang H, Zafar MH, Wang M. Research progress in the role and mechanism of Leucine in regulating animal growth and development. Front Physiol 2023; 14:1252089. [PMID: 38046946 PMCID: PMC10691278 DOI: 10.3389/fphys.2023.1252089] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/08/2023] [Indexed: 12/05/2023] Open
Abstract
Leucine, a branched-chain amino acid, is essential in regulating animal growth and development. Recent research has uncovered the mechanisms underlying Leucine's anabolic effects on muscle and other tissues, including its ability to stimulate protein synthesis by activating the mTORC1 signaling pathway. The co-ingestion of carbohydrates and essential amino acids enhances Leucine's anabolic effects. Moreover, Leucine has been shown to benefit lipid metabolism, and insulin sensitivity, making it a promising strategy for preventing and treating metabolic diseases, including type 2 diabetes and obesity. While emerging evidence indicates that epigenetic mechanisms may mediate Leucine's effects on growth and development, more research is needed to elucidate its mechanisms of action fully. Specific studies have demonstrated that Leucine promotes muscle growth and metabolic health in animals and humans, making it a promising therapeutic agent. However, it is essential to note that Leucine supplementation may cause digestive issues or interact with certain medications, and More study is required to determine definitively optimal dosages. Therefore, it is important to understand how Leucine interacts with other nutrients, dietary factors, and lifestyle habits to maximize its benefits. Overall, Leucine's importance in human nutrition is far-reaching, and its potential to prevent muscle loss and enhance athletic performance warrants further investigation.
Collapse
Affiliation(s)
| | | | | | | | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
44
|
Franklin IG, Milne P, Childs J, Boggan RM, Barrow I, Lawless C, Gorman GS, Ng YS, Collin M, Russell OM, Pickett SJ. T cell differentiation drives the negative selection of pathogenic mitochondrial DNA variants. Life Sci Alliance 2023; 6:e202302271. [PMID: 37652671 PMCID: PMC10471888 DOI: 10.26508/lsa.202302271] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 09/02/2023] Open
Abstract
Pathogenic mitochondrial DNA (mtDNA) single-nucleotide variants are a common cause of adult mitochondrial disease. Levels of some variants decrease with age in blood. Given differing division rates, longevity, and energetic requirements within haematopoietic lineages, we hypothesised that cell-type-specific metabolic requirements drive this decline. We coupled cell-sorting with mtDNA sequencing to investigate mtDNA variant levels within progenitor, myeloid, and lymphoid lineages from 26 individuals harbouring one of two pathogenic mtDNA variants (m.3243A>G and m.8344A>G). For both variants, cells of the T cell lineage show an enhanced decline. High-throughput single-cell analysis revealed that decline is driven by increasing proportions of cells that have cleared the variant, following a hierarchy that follows the current orthodoxy of T cell differentiation and maturation. Furthermore, patients with pathogenic mtDNA variants have a lower proportion of T cells than controls, indicating a key role for mitochondrial function in T cell homeostasis. This work identifies the ability of T cell subtypes to selectively purify their mitochondrial genomes, and identifies pathogenic mtDNA variants as a new means to track blood cell differentiation status.
Collapse
Affiliation(s)
- Imogen G Franklin
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, England
| | - Paul Milne
- Haematopoiesis and Immunogenomics Laboratory, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, England
| | - Jordan Childs
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, England
| | - Róisín M Boggan
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, England
| | - Isabel Barrow
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, England
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, England
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, England
| | - Conor Lawless
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, England
| | - Gráinne S Gorman
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, England
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, England
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, England
| | - Yi Shiau Ng
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, England
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, England
| | - Matthew Collin
- Haematopoiesis and Immunogenomics Laboratory, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, England
| | - Oliver M Russell
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, England
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, England
| | - Sarah J Pickett
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, England
| |
Collapse
|
45
|
Xiao J, Chen X, Liu W, Qian W, Bulek K, Hong L, Miller-Little W, Li X, Liu C. TRAF4 is crucial for ST2+ memory Th2 cell expansion in IL-33-driven airway inflammation. JCI Insight 2023; 8:e169736. [PMID: 37607012 PMCID: PMC10561728 DOI: 10.1172/jci.insight.169736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 08/17/2023] [Indexed: 08/23/2023] Open
Abstract
Tumor necrosis factor receptor-associated factor 4 (TRAF4) is an important regulator of type 2 responses in the airway; however, the underlying cellular and molecular mechanisms remain elusive. Herein, we generated T cell-specific TRAF4-deficient (CD4-cre Traf4fl/fl) mice and investigated the role of TRAF4 in memory Th2 cells expressing IL-33 receptor (ST2, suppression of tumorigenicity 2) (ST2+ mTh2 cells) in IL-33-mediated type 2 airway inflammation. We found that in vitro-polarized TRAF4-deficient (CD4-cre Traf4fl/fl) ST2+ mTh2 cells exhibited decreased IL-33-induced proliferation as compared with TRAF4-sufficient (Traf4fl/fl) cells. Moreover, CD4-cre Traf4fl/fl mice showed less ST2+ mTh2 cell proliferation and eosinophilic infiltration in the lungs than Traf4fl/fl mice in the preclinical models of IL-33-mediated type 2 airway inflammation. Mechanistically, we discovered that TRAF4 was required for the activation of AKT/mTOR and ERK1/2 signaling pathways as well as the expression of transcription factor Myc and nutrient transporters (Slc2a1, Slc7a1, and Slc7a5), signature genes involved in T cell growth and proliferation, in ST2+ mTh2 cells stimulated by IL-33. Taken together, the current study reveals a role of TRAF4 in ST2+ mTh2 cells in IL-33-mediated type 2 pulmonary inflammation, opening up avenues for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Jianxin Xiao
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Xing Chen
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Weiwei Liu
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Wen Qian
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Katarzyna Bulek
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Lingzi Hong
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - William Miller-Little
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
- Medical Scientist Training Program
- Department of Pathology, and
| | - Xiaoxia Li
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Caini Liu
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
46
|
Kristian YY, Cahyanur R, Wulandari Y, Sinaga W, Lukito W, Prasetyawaty F, Lestari W. Correlation between branched-chain amino acids intake and total lymphocyte count in head and neck cancer patients: a cross-sectional study. BMC Nutr 2023; 9:86. [PMID: 37452428 PMCID: PMC10347797 DOI: 10.1186/s40795-023-00746-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Cellular immunity as reflected by total lymphocyte count (TLC) has been proven to be related to overall survival rate cancer patients. Lymphocyte proliferation is regulated, to some extent, by nutritional factor. Branched chain amino acid (BCAA) is documented as one of numerous nutrients that play important role in lymphocyte proliferation through its effect on protein synthesis and DNA replication. Many studies describe the correlation between BCAA and TLC in hepatic cancer patients. This study emphasized the observation of that links in head and neck cancer patients. METHODS Eighty-five subjects were included in final analysis, aged 18-75, mostly male, with head and neck cancer who had not received treatment participated in this cross-sectional study at the Dr. Cipto Mangunkusumo General Hospital's radiation and medical haematology oncology clinic. The BCAAs intake was assessed using a semi-quantitative food frequency questionnaire. Flow cytometry method was used to quantify TLC. RESULTS Overall, the subjects' nutritional status mostly was considered normal, with the median intake of 1505 (800-3040) kcal/day of energy and mean of 73.96 ± 23.39 g/day of protein. Moreover, subjects' average BCAA intake was 10.92 ± 0.48 g/day. Meanwhile, 17.6% of subjects were found to have low TLC level. From thorough analysis, we did not find a strong correlation between BCAA level and TLC (r = 0.235, p = 0.056). CONCLUSION In participants with head and neck cancer who had not received chemoradiotherapy, there is no correlation between BCAA intake and TLC. The contribution of non-BCAA amino acids from dietary sources to lymphocyte proliferation requires further investigation. TRIAL REGISTRATION Retrospectively registered, with clinical trial number NCT05226065 on February 7th 2022.
Collapse
Affiliation(s)
- Yosua Yan Kristian
- Department of Nutrition, Faculty of Medicine, Universitas Indonesia - Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia.
| | - Rahmat Cahyanur
- Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia - Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Yohannessa Wulandari
- Department of Nutrition, Faculty of Medicine, Universitas Indonesia - Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Wina Sinaga
- Department of Nutrition, Faculty of Medicine, Universitas Indonesia - Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Widjaja Lukito
- Department of Nutrition, Faculty of Medicine, Universitas Indonesia - Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Findy Prasetyawaty
- Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia - Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Wiji Lestari
- Department of Nutrition, Faculty of Medicine, Universitas Indonesia - Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| |
Collapse
|
47
|
Cheng J, Yan J, Liu Y, Shi J, Wang H, Zhou H, Zhou Y, Zhang T, Zhao L, Meng X, Gong H, Zhang X, Zhu H, Jiang P. Cancer-cell-derived fumarate suppresses the anti-tumor capacity of CD8 + T cells in the tumor microenvironment. Cell Metab 2023:S1550-4131(23)00171-7. [PMID: 37178684 DOI: 10.1016/j.cmet.2023.04.017] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 03/06/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023]
Abstract
Metabolic alterations in the microenvironment significantly modulate tumor immunosensitivity, but the underlying mechanisms remain obscure. Here, we report that tumors depleted of fumarate hydratase (FH) exhibit inhibition of functional CD8+ T cell activation, expansion, and efficacy, with enhanced malignant proliferative capacity. Mechanistically, FH depletion in tumor cells accumulates fumarate in the tumor interstitial fluid, and increased fumarate can directly succinate ZAP70 at C96 and C102 and abrogate its activity in infiltrating CD8+ T cells, resulting in suppressed CD8+ T cell activation and anti-tumor immune responses in vitro and in vivo. Additionally, fumarate depletion by increasing FH expression strongly enhances the anti-tumor efficacy of anti-CD19 CAR T cells. Thus, these findings demonstrate a role for fumarate in controlling TCR signaling and suggest that fumarate accumulation in the tumor microenvironment (TME) is a metabolic barrier to CD8+ T cell anti-tumor function. And potentially, fumarate depletion could be an important strategy for tumor immunotherapy.
Collapse
Affiliation(s)
- Jie Cheng
- School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jinxin Yan
- School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ying Liu
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jiangzhou Shi
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei 430081, China
| | - Haoyu Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Hanyang Zhou
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yinglin Zhou
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Tongcun Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei 430081, China
| | - Lina Zhao
- School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Xianbin Meng
- National Center for Protein Science, Tsinghua University, Beijing 100084, China
| | - Haipeng Gong
- School of Life Sciences, Tsinghua University, Beijing 100084, China; MOE Key Laboratory of Bioinformatics, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xinxiang Zhang
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Haichuan Zhu
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei 430081, China.
| | - Peng Jiang
- School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
48
|
Bremova-Ertl T, Schneider S. Current advancements in therapy for Niemann-Pick disease: progress and pitfalls. Expert Opin Pharmacother 2023; 24:1229-1247. [PMID: 37211769 DOI: 10.1080/14656566.2023.2215386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
INTRODUCTION Niemann-Pick disease type C (NPC) is a rare, autosomal recessive, lysosomal storage disorder. To combat the progressive neurodegeneration in NPC, disease-modifying treatment needs to be introduced early in the course of the disease. The only approved, disease-modifying treatment is a substrate-reduction treatment, miglustat. Given miglustat's limited efficacy, new compounds are under development, including gene therapy; however, many are still far from clinical use. Moreover, the phenotypic heterogeneity and variable course of the disease can impede the development and approval of new agents. AREAS COVERED Here, we offer an expert review of these therapeutic candidates, with a broad scope not only on the main pharmacotherapies, but also on experimental approaches, gene therapies, and symptomatic strategies. The National Institute of Health (NIH) database PubMed has been searched for the combination of the words 'Niemann-Pick type C'+ 'treatment' or 'therapy' or 'trial.' The website clinicaltrials.gov has also been consulted. EXPERT OPINION We conclude a combination of treatment strategies should be sought, with a holistic approach, to improve the quality of life of affected individuals and their families.
Collapse
Affiliation(s)
- Tatiana Bremova-Ertl
- Department of Neurology, University Hospital Bern (Inselspital) and University of Bern, Bern, Switzerland
- Center for Rare Diseases, University Hospital Bern (Inselspital) and University of Bern, Bern, Switzerland
| | - Susanne Schneider
- Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
49
|
Ganjoo S, Gupta P, Corbali HI, Nanez S, Riad TS, Duong LK, Barsoumian HB, Masrorpour F, Jiang H, Welsh JW, Cortez MA. The role of tumor metabolism in modulating T-Cell activity and in optimizing immunotherapy. Front Immunol 2023; 14:1172931. [PMID: 37180129 PMCID: PMC10169689 DOI: 10.3389/fimmu.2023.1172931] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
Immunotherapy has revolutionized cancer treatment and revitalized efforts to harness the power of the immune system to combat a variety of cancer types more effectively. However, low clinical response rates and differences in outcomes due to variations in the immune landscape among patients with cancer continue to be major limitations to immunotherapy. Recent efforts to improve responses to immunotherapy have focused on targeting cellular metabolism, as the metabolic characteristics of cancer cells can directly influence the activity and metabolism of immune cells, particularly T cells. Although the metabolic pathways of various cancer cells and T cells have been extensively reviewed, the intersections among these pathways, and their potential use as targets for improving responses to immune-checkpoint blockade therapies, are not completely understood. This review focuses on the interplay between tumor metabolites and T-cell dysfunction as well as the relationship between several T-cell metabolic patterns and T-cell activity/function in tumor immunology. Understanding these relationships could offer new avenues for improving responses to immunotherapy on a metabolic basis.
Collapse
Affiliation(s)
- Shonik Ganjoo
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Priti Gupta
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Halil Ibrahim Corbali
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Selene Nanez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Thomas S. Riad
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lisa K. Duong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hampartsoum B. Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Fatemeh Masrorpour
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hong Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James W. Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria Angelica Cortez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
50
|
Zhao Y, Li F, Ma F, Zhi J, Wu G, Zheng X. Theoretical prediction of nanomolar and sequence-selective binding of synthetic supramolecular cucurbit[7]uril to N-terminal Leu-containing tripeptides. Phys Chem Chem Phys 2023; 25:7893-7900. [PMID: 36857719 DOI: 10.1039/d2cp03818h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Molecular recognition towards peptides and proteins with high affinity by synthetic supramolecular hosts is important but challenging. In this work, we investigate the molecular recognition of the synthetic cucurbit[7]uril (CB[7]) to 17 designed N-terminal Leu-containing tripeptides in aqueous medium by molecular dynamics (MD) simulation and screen out tripeptides with high binding affinity. It is found that, compared to LGG, only the third residue is Arg (R), the binding affinity of CB[7] to LGR reaches nanomolar level with binding equilibrium constant (Ka) of 1.1 × 109 M-1. The CB[7] recognition to the N-terminal Leu-containing tripeptides is highly sequence dependent; whether changing the sequence order (from LGR to LRG) or increasing the sequence length (from LGR to LGGR), Ka decreases by about three orders of magnitude. Interestingly, substituting N-terminal Leu for its isomer Ile, the binding of CB[7] to tripeptides weakens significantly with Ka decreasing by 3-8 orders of magnitude. Thus CB[7] can effectively distinguish N-terminal Leu-containing tripeptides from N-terminal Ile-containing tripeptides. Importantly, we predict that when R is as C-terminus, regardless of N-terminal residue being of aromatic type or Leu, the binding strength is always close to the nanomolar level. Therefore, R can be introduced to rationally design novel peptides with high binding affinity to CB[7] in practical applications.
Collapse
Affiliation(s)
- Ying Zhao
- Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, China.
| | - Fei Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, China.
| | - Fenfen Ma
- GuSu Laboratory of Materials, Suzhou 215123, Jiangsu, China
| | - Junge Zhi
- Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, China.
| | - Guanglu Wu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, China.
| | - Xiaoyan Zheng
- Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, China. .,Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates (South China University of Technology), Guangzhou 510640, China
| |
Collapse
|