1
|
Xie J, Zheng Z, Wang B, Zhang J, Jiang J, Wu F, Zhong X, Chen J. LncRNA HOTAIR promotes aerobic glycolysis by recruiting Lin28 to induce inflammation and apoptosis in acute lung injury. RNA Biol 2025; 22:1-12. [PMID: 40052944 PMCID: PMC11901367 DOI: 10.1080/15476286.2025.2475255] [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] [Revised: 01/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Acute lung injury (ALI) is a life-threatening condition with high rates of morbidity and mortality. Recently, there has been growing evidence suggesting a link between lncRNA HOTAIR and ALI. Nonetheless, the precise role and mechanism of lncRNA HOTAIR in ALI remain to be fully elucidated. siHOTAIR transfection, qPCR detection (HOTAIR), ELISA (TNF-α, IL-6, and IL-1β), Lactate detection, Glucose uptake experiment, Cell Apoptosis Analysis, Fluorescence in situ hybridization (FISH) assay. Through siHOTAIR transfection, we discovered that HOTAIR plays a role in the secretion of inflammatory factors in ALI and further regulates glucose uptake and metabolism in lung epithelial cells. Moreover, a comparison between HOTAIR knockdown cells and HOTAIR overexpression cells revealed that HOTAIR promotes cellular aerobic sugar metabolism, leading to increased secretion of inflammatory factors and cell apoptosis. Our in-depth research also identified an interaction between HOTAIR and the LIN28 protein. Knocking down HOTAIR resulted in the downregulation of LIN28 protein expression, which subsequently inhibited the expression of the glucose transporter GLUT1. This indicates that HOTAIR facilitates glucose uptake and boosts cellular aerobic glycolysis by modulating the LIN28 protein, thereby promoting inflammation and apoptosis in acute lung injury. The research findings presented in this article offer significant insights into the function of HOTAIR in ALI and suggest a potential therapeutic target for the treatment of this condition.
Collapse
Affiliation(s)
- Junjie Xie
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Zhicong Zheng
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Bin Wang
- Department of Pediatrics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jianfang Zhang
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Junqi Jiang
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Fengde Wu
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Xiangming Zhong
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Jianfeng Chen
- Department of Pediatrics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Yang Q, Guo C, Zhang L. The role of metabolite sensors in metabolism-immune interaction: New targets for immune modulation. Clin Transl Med 2025; 15:e70294. [PMID: 40159576 DOI: 10.1002/ctm2.70294] [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: 03/17/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025] Open
Abstract
Recent advancements in immunometabolism have highlighted the critical role of metabolite sensors in regulating immune responses. Metabolites such as lactate, succinate, itaconate, and β-hydroxybutyrate influence immune cell function by interacting with specific sensors. These metabolites act as signaling molecules, linking cellular metabolic changes to immune responses. Lactate, a metabolite commonly produced under hypoxic conditions, has emerged as a major regulator of innate immunity. Key enzymes, including AARS1 and AARS2, function as intracellular lactate sensors, catalyzing lactylation on proteins like cGAS, which plays a central role in DNA sensing and immune activation. The lactylation of cGAS inhibits its activity, modulating immune responses by balancing inflammation and immune tolerance. Metabolite sensors, like MCT1, also contribute to immune modulation, particularly in cancer and chronic inflammatory diseases. Therapeutically, targeting these sensors offers potential for restoring immune function, especially in cancer immunotherapy. However, challenges in specificity, off-target effects, and long-term safety require further investigation. This article explores the emerging role of metabolite sensors in immune regulation, with a focus on lactate sensors, and outlines potential therapeutic strategies to enhance immune responses in metabolic diseases.
Collapse
Affiliation(s)
- Qiqing Yang
- Department of Radiation Oncology and the State Key Laboratory of Transvascular Implantation Devices, the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Ce Guo
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University School of Medicine, Hangzhou, China
- The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Deng L, Wang J, Deng Y, Huang J, Gu Q, Chen Q, Pan L, Wei J, Wang Q, Sun L. Effect of dapagliflozin on malignant ventricular arrhythmias in elderly after acute myocardial infarction: a propensity score-matched cohort study. Eur J Clin Pharmacol 2025:10.1007/s00228-025-03832-8. [PMID: 40167624 DOI: 10.1007/s00228-025-03832-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVE This study aims to evaluate the effect of dapagliflozin (DAPA) on malignant ventricular arrhythmias (MVA) after acute myocardial infarction (AMI). METHODS A single-center, prospective and observational cohort study was conducted. We enrolled AMI patients from the ChangZhou Acute Myocardial Infarction Registry between January 2018 and November 2023. They were divided into two groups according to the use of dapagliflozin. The median follow-up time was 211 days. The primary endpoint of the study was the incidence of MVA during hospitalization, and the secondary endpoint was all-cause mortality rate during the follow-up period. Kaplan-Meier survival analysis and multifactorial logistic regression analysis were performed to assess the association between DAPA and the risk of MVA. Enrolled patients were matched on a 1:1 propensity score. RESULTS Of the 2607 AMI patients enrolled, MVA were reported postoperatively in 123 (4.7%)patients. Cardiovascular death occurred in 93 (3.6%) patients. The average age of the enrolled patients was 65.03 ± 0.27 years. Of participants assigned to dapagliflozin, 8 out of 363 patients (2.2%) experienced MVA compared with 115 out of 2244 patients (5.1%) in the control group (odds ratio, OR = 0.392; 95% confidence interval, 95% CI: 0.171-0.900; P = 0.027). After 1:1 propensity score matching, DAPA remained able to reduce the risk of MVA in patients with AMI. (OR = 0.340; 95% CI: 0.121-0.960; P = 0.042). At a median follow-up of 211 days, all-cause mortality remained lower in the DAPA group than in the control group after matching (P = 0.033). CONCLUSION Dapagliflozin may attenuate the risk of MVA and all-cause mortality in elderly AMI patients, highlighting its potential as a therapeutic adjunct. However, these findings require validation in large-scale randomized trials.
Collapse
Affiliation(s)
- Li Deng
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214001, Jiangsu, China
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Jingyi Wang
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Ye Deng
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Jianya Huang
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Qingqing Gu
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Qianwen Chen
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Lu Pan
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214001, Jiangsu, China
| | - Jun Wei
- Department of Cardiovascular Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China.
| | - Qingjie Wang
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China.
| | - Ling Sun
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214001, Jiangsu, China.
| |
Collapse
|
4
|
El-Sehrawy AAMA, Ayoub II, Uthirapathy S, Ballal S, Gabble BC, Singh A, V K, Panigrahi R, Kamali M, Khosravi M. The microbiota-gut-brain axis in myalgic encephalomyelitis/chronic fatigue syndrome: a narrative review of an emerging field. Eur J Transl Myol 2025; 35. [PMID: 39937103 DOI: 10.4081/ejtm.2025.13690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
The intricate relationship between gut microbiota and the brain has emerged as a pivotal area of research, particularly in understanding Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). This complex condition is characterized by debilitating fatigue, cognitive dysfunction, and a wide array of systemic manifestations, posing significant challenges for diagnosis and treatment. Recent studies highlight the microbiota-gut-brain axis as a crucial pathway in ME/CFS pathophysiology, suggesting that alterations in gut microbial composition may impact immune responses, neurochemical signaling, and neuronal health. This narrative review systematically explores English-language scholarly articles from January 1995 to January 2025, utilizing databases such as PubMed, Scopus, and Web of Science. The findings underscore the potential for targeted therapeutic interventions aimed at correcting gut dysbiosis. As research progresses, a deeper understanding of the microbiota-gut-brain connection could lead to innovative approaches for managing ME/CFS, ultimately enhancing the quality of life for affected individuals.
Collapse
Affiliation(s)
| | | | - Subasini Uthirapathy
- Faculty of Pharmacy, Department of Pharmacology, Tishk International University, Erbil, Kurdistan Region.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka.
| | - Baneen C Gabble
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon.
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab.
| | - Kavitha V
- Department of Chemistry, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu.
| | - Rajashree Panigrahi
- Department of Microbiology, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar.
| | - Mostafa Kamali
- Department of Psychiatry, School of Medicine, Zahedan University of Medical Sciences, Zahedan.
| | - Mohsen Khosravi
- Department of Psychiatry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran; Health Promotion Research Center, Zahedan University of Medical Sciences, Zahedan, Iran; Community Nursing Research Center, Zahedan University of Medical Sciences, Zahedan.
| |
Collapse
|
5
|
Wang C, Xu W, Jiang S, Wu Y, Shu J, Gao X, Huang K. β-Hydroxybutyrate Facilitates Postinfarction Cardiac Repair via Targeting PHD2. Circ Res 2025; 136:704-718. [PMID: 39957619 DOI: 10.1161/circresaha.124.325179] [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: 07/04/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Acute myocardial infarction (MI) remains one of the major causes of death worldwide, and innovative treatment strategies for MI represent a major challenge in cardiovascular medicine. Caloric restriction (CR) is the most potent nonpharmacological intervention known to prevent age-related disorders and extend lifespan. CR reduces glycolysis and elevates ketone body metabolism. However, whether and how CR or ketone body prevents the progression of MI remains poorly defined. METHODS Mice treated with CR and β-hydroxybutyrate (β-OHB) underwent MI induced by ligation of the left anterior descending coronary artery. Cardiac function was assessed by echocardiographic measurements. Histological analysis, fluorescence-activated cell sorting, and immunofluorescence were used to assess myocardial neovascularization and macrophage filtration. The interaction and modification of β-OHB on PHD2 were analyzed by molecular docking, cellular thermal shift assay, liquid chromatography with tandem mass spectrometry, and coimmunoprecipitation. Macrophage-specific PHD2 K239R and K385R knock-in mice were used to determine the functional significance of β-OHB/PHD2 axis in vivo. RESULTS Twelve weeks of CR markedly rescued postinfarction cardiac function by enhancing neovascularization. CR significantly increased circulating and cardiac ketone bodies, including β-OHB and acetoacetate. We identified β-OHB but not acetoacetate selectively targeted macrophages to stimulate VEGF (vascular endothelial growth factor) production in the peri-infarct area to promote neovascularization and cardiac repair. Mechanistically, β-OHB binds to and induces lysine β-hydroxybutyrylation of PHD2 at lysines 239 and 385, thus blocking its function in the hydroxylation of HIF-1α (hypoxia-inducible factor 1α) and resulting in enhanced HIF1α-dependent VEGF transcription and secretion. More importantly, specific PHD2 lys239 and lys385 mutations in macrophages abolished the preventive effects of exogenous β-OHB on MI in mice. CONCLUSIONS These data reveal a novel regulation of lysine β-hydroxybutyrylation on PHD2 and demonstrate a promising and therapeutic role for β-OHB/PHD2 in effectively accelerating neovascularization and preserving heart function after cardiac ischemia.
Collapse
Affiliation(s)
- Cheng Wang
- Clinic Center of Human Gene Research, Union Hospital (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Rheumatology and Immunology, Union Hospital (C.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital (W.X., S.J., Y.W., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjing Xu
- Clinic Center of Human Gene Research, Union Hospital (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital (W.X., S.J., Y.W., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shushu Jiang
- Clinic Center of Human Gene Research, Union Hospital (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital (W.X., S.J., Y.W., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yichen Wu
- Clinic Center of Human Gene Research, Union Hospital (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital (W.X., S.J., Y.W., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiangcheng Shu
- Clinic Center of Human Gene Research, Union Hospital (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyuan Gao
- Clinic Center of Human Gene Research, Union Hospital (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital (W.X., S.J., Y.W., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Huang
- Clinic Center of Human Gene Research, Union Hospital (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital (W.X., S.J., Y.W., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging (C.W., W.X., S.J., Y.W., J.S., X.G., K.H.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Ruan Z, Wang Y, Shi L, Yang XJ. Progress of research on glucose transporter proteins in hepatocellular carcinoma. World J Hepatol 2025; 17:104715. [DOI: 10.4254/wjh.v17.i3.104715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/02/2025] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumour with high prevalence and mortality rate worldwide. Metabolic reprogramming of cancer cells may be a major factor in the process of this disease. Glucose transporter proteins (GLUTs) are members of the major facilitator superfamily of membrane transporters, playing a pivotal role in the metabolic reprogramming and tumour progression in HCC. This review discusses the advances in the study of GLUTs in HCC, including the expression patterns, functions and possibilities of GLUTs. In HCC, the expression levels of GLUTs are closely associated with tumour aggressiveness, metabolic reprogramming and prognosis. A series of inhibitors have been demonstrated efficacy in inhibiting HCC cell growth and glucose uptake in in vitro and in vivo models. These inhibitors offer a novel approach to HCC treatment by reducing the glucose metabolism of tumour cells, thereby impeding tumour growth, and concurrently enhancing the sensitivity to chemotherapeutic agents. This reminds us of the urgent need to elucidate GLUTs’ roles in HCC and to determine the most effective ways to translate these findings into clinical practice.
Collapse
Affiliation(s)
- Zheng Ruan
- The First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Yan Wang
- Division of Personnel, Gansu Provincial People’s Hospital, Lanzhou 730000, Gansu Province, China
| | - Lei Shi
- Department of General Surgery, The Second people’s Hospital of Lanzhou, Lanzhou 730000, Gansu Province, China
| | - Xiao-Jun Yang
- Department of General Surgery, Gansu Provincial People’s Hospital, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
7
|
Tsusaka T, Najar MA, Schwarz B, Bohrnsen E, Oses-Prieto JA, Neudorf H, Lee C, Little JP, Burlingame AL, Bosio CM, Burslem GM, Goldberg EL. Reversible histone deacetylase activity catalyzes lysine acylation. Nat Chem Biol 2025:10.1038/s41589-025-01869-5. [PMID: 40140626 DOI: 10.1038/s41589-025-01869-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 02/27/2025] [Indexed: 03/28/2025]
Abstract
The dynamic modification of proteins by many metabolites suggests an intimate link between energy metabolism and post-translational modifications (PTMs). For instance, starvation and low-carbohydrate diets lead to the accumulation of β-hydroxybutyrate (BHB), whose blood concentrations can reach millimolar levels, concomitant with the accumulation of lysine β-hydroxybutyrylation (Kbhb) of proteins. Here we report that class I histone deacetylases (HDACs) unexpectedly catalyze the formation of Kbhb. Through mutational analysis, we show a shared reliance on key active site amino acids for classical deacetylation and noncanonical HDAC-catalyzed β-hydroxybutyrylation. On the basis of these data, we propose that HDACs catalyze a condensation reaction between the free amine group on lysine and the BHB carboxylic acid, thereby generating an amide bond. This reversible HDAC activity is not limited to BHB and extends to multiple short-chain fatty acids, representing a novel mechanism of PTM deposition relevant to metabolically sensitive proteome modifications.
Collapse
Affiliation(s)
- Takeshi Tsusaka
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Mohd Altaf Najar
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin Schwarz
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| | - Eric Bohrnsen
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Helena Neudorf
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia, Canada
| | - Christina Lee
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Jonathan P Little
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia, Canada
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Catharine M Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| | - George M Burslem
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cancer Biology and Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily L Goldberg
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
8
|
Yan J, Han VX, Jones HF, Couttas TA, Jieu B, Leweke FM, Lee J, Loi C, Webster R, Kothur K, Menezes MP, Antony J, Kandula T, Cardamone M, Patel S, Bandodkar S, Dale RC. Cerebrospinal fluid metabolomics in autistic regression reveals dysregulation of sphingolipids and decreased β-hydroxybutyrate. EBioMedicine 2025; 114:105664. [PMID: 40138886 DOI: 10.1016/j.ebiom.2025.105664] [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: 11/20/2024] [Revised: 03/05/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Autism is highly heritable, however actionable genetic findings are only found in a minority of patients. Many people with autism suffer loss of neurodevelopmental skills, known as autistic regression. The cause of regression is poorly understood, and the diagnostic and therapeutic pathways are lacking. METHODS We used untargeted metabolomics using a UPLC-Q-Exactive-HFx Mass Spectrometry to examine cerebrospinal fluid (CSF) from twenty-two patients with autistic regression compared to sixteen controls with neurodevelopmental disorders (but not autistic regression) and thirty-four controls with other neurological disease (headache, encephalitis, epilepsy). The twenty-two patients with autistic regression consisted of two groups: early (infantile) autistic regression <2 years of age (n = 8), and later regression of skills >4 years of age, often in the context of pre-existing developmental concerns (n = 14). Metabolites of interest were then quantified and validated using targeted assays. FINDINGS Untargeted case-control studies revealed good separation of patients from controls using multivariate analysis. β-hydroxybutyrate was significantly decreased in the CSF of patients with autistic regression, and the findings were validated using a targeted β-hydroxybutyrate assay. The sphingolipid, sphingosine-1-phosphate was significantly elevated in the discovery case-control studies, and sphingolipid metabolism pathways were also significantly dysregulated. We therefore developed a targeted metabolite assay of forty sphingolipids. After FDR correction, 21 of the 40 sphingolipids were significantly dysregulated (pFDR < 0.05) (Benjamini-Hochberg correction) in autistic regression compared to the neurodevelopmental controls, and 26 of the 40 sphingolipids were significantly dysregulated in autistic regression compared to other neurological controls, with elevated ceramides, hexosylceramides, sphingosines (including sphingosine-1-phosphate), and sulfatides. By contrast, sphingomyelin levels were generally decreased in autistic regression. INTERPRETATION Our data shows the potential utility of CSF metabolomics in the context of autistic regression, a clinical syndrome which has historically lacked pathophysiological biomarkers and disease modifying therapies. FUNDING Financial support for the study was granted by Dale NHMRC Investigator grant APP1193648, Petre Foundation, Cerebral Palsy Alliance, and Ainsworth and SCHF Neuroscience grant scheme.
Collapse
Affiliation(s)
- Jingya Yan
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Velda X Han
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Hannah F Jones
- Starship Hospital, Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Timothy A Couttas
- Neuroscience Research Australia, Randwick, NSW, Australia; Brain and Mind Centre, The University of Sydney, NSW, Australia
| | - Beverly Jieu
- Brain and Mind Centre, The University of Sydney, NSW, Australia
| | - F Markus Leweke
- Brain and Mind Centre, The University of Sydney, NSW, Australia
| | - Jennifer Lee
- Department of Endocrinology, The Children's Hospital at Westmead, NSW, Australia
| | - Catherine Loi
- Department of Endocrinology, The Children's Hospital at Westmead, NSW, Australia
| | - Richard Webster
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Kavitha Kothur
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Manoj P Menezes
- Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Jayne Antony
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Tejaswi Kandula
- Department of Neurology, Sydney Children's Hospital Network, Sydney, NSW, Australia
| | - Michael Cardamone
- Department of Neurology, Sydney Children's Hospital Network, Sydney, NSW, Australia
| | - Shrujna Patel
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Sushil Bandodkar
- Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Department of Biochemistry, The Children's Hospital at Westmead, NSW, Australia
| | - Russell C Dale
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia.
| |
Collapse
|
9
|
Flower L, Vozza EG, Bryant CE, Summers C. Role of inflammasomes in acute respiratory distress syndrome. Thorax 2025; 80:255-263. [PMID: 39884849 DOI: 10.1136/thorax-2024-222596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/10/2025] [Indexed: 02/01/2025]
Abstract
Acute respiratory distress syndrome (ARDS) is present in >10% of all people admitted to critical care and is associated with severe morbidity and mortality. Despite more than half a century since its first description, no efficacious pharmacological therapies have been developed, and little progress has been made in improving clinical outcomes. Neutrophils are the principal drivers of ARDS, with their priming and subsequent aberrant downstream functions, including interleukin (IL) 1β and IL-18 secretion, central to the disease pathogenesis. The dominant pathways through which IL-1β and IL-18 are believed to be elaborated are multimeric protein structures called inflammasomes that consist of sensor proteins, adaptor proteins and an effector enzyme. The inflammasome's initial activation depends on one of a variety of damage-associated (DAMP) or pathogen-associated (PAMP) molecular patterns. However, once activated, a common downstream inflammatory pathway is initiated regardless of the specific DAMP or PAMP involved. Several inflammasomes exist in humans. The nucleotide-binding domain leucine-rich repeat (NLR) family, pyrin domain-containing 3 (NLRP3), inflammasome is the best described in the context of ARDS and is known to be activated in both infective and sterile cases. The NLR family, caspase activation and recruitment domain-containing 4 (NLRC4) and absent in melanoma 2 (AIM2) inflammasomes have also been implicated in various ARDS settings, as have inflammasome-independent pathways. Further work is required to understand human biology as much of our knowledge is extrapolated from rodent experimental models. Experimental lung injury models have demonstrated beneficial responses to inflammasome, IL-1β and IL-18 blockade. However, findings have yet to be successfully translated into humans with ARDS, likely due to an underappreciation of the central role of the neutrophil inflammasome. A thorough understanding of inflammasome pathways is vital for critical care clinicians and researchers and for the development of beneficial therapies. In this review, we describe the central role of the inflammasome in the development of ARDS and its potential for immunomodulation, highlighting key areas for future research.
Collapse
Affiliation(s)
- Luke Flower
- Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Emilio G Vozza
- Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Clare E Bryant
- Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Charlotte Summers
- Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
10
|
Qiu X, Wu W, Zhang S, Huang C, Lin D. 3-Hydroxybutyrate Promotes Myoblast Proliferation and Differentiation through Energy Metabolism and GPR109a-Mediated Ca 2+-NFAT Signaling Pathways. J Proteome Res 2025. [PMID: 40099866 DOI: 10.1021/acs.jproteome.4c01150] [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: 03/20/2025]
Abstract
Skeletal muscle wasting is a critical clinical problem associated with several diseases that significantly impair patient outcomes due to the progressive loss of muscle mass and function. This study explores the potential of 3-hydroxybutyrate (3-HB) as a therapeutic agent to counteract muscle atrophy by promoting the proliferation and differentiation of C2C12 myoblasts. Using nuclear magnetic resonance (NMR)-based metabolomics analysis, we uncover the underlying mechanisms by which 3-HB exerts its effects. Our findings demonstrate that 3-HB exerts its effects through two distinct mechanisms: as a metabolic substrate and as a signaling molecule. As a metabolic substrate, 3-HB enhances myoblast energy efficiency by stimulating the expression of G protein-coupled receptor 109a (GPR109a), which subsequently upregulates the 3-HB transporters MCT1 and CD147, the utilization enzyme OXCT1, and phosphorylated AMPK, thereby increasing ATP production. As a signaling molecule, 3-HB activates GPR109a, promoting calcium influx, improving calcium homeostasis, and increasing the expression of Ca2+-related proteins such as CAMKK2. This signaling cascade activates calcineurin (CaN), facilitating NFAT translocation to the nucleus and gene expression that drives myoblast proliferation and differentiation. By elucidating the dual regulatory roles of 3-HB in energy metabolism and cellular signaling, this study not only advances our understanding of muscle physiology but also highlights the potential of 3-HB as a novel therapeutic approach for the prevention or treatment of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Xu Qiu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Wenfang Wu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Shuya Zhang
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Caihua Huang
- Research and Communication Center of Exercise and Health, Xiamen University of Technology, Xiamen 361024, China
| | - Donghai Lin
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
11
|
Nabakhteh S, Lotfi A, Afsartaha A, Khodadadi ES, Abdolghaderi S, Mohammadpour M, Shokri Y, Kiani P, Ehtiati S, Khakshournia S, Khatami SH. Nutritional Interventions in Amyotrophic Lateral Sclerosis: From Ketogenic Diet and Neuroprotective Nutrients to the Microbiota-Gut-Brain Axis Regulation. Mol Neurobiol 2025:10.1007/s12035-025-04830-8. [PMID: 40097762 DOI: 10.1007/s12035-025-04830-8] [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: 10/04/2024] [Accepted: 03/09/2025] [Indexed: 03/19/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex neurodegenerative disease with significant challenges in diagnosis and treatment. Recent research has highlighted the complex nature of ALS, encompassing behavioral impairments in addition to its neurological manifestations. While several medications have been approved to slow disease progression, ongoing research is focused on identifying new therapeutic targets. The current review focuses on emerging therapeutic strategies and personalized approaches aimed at improving patient outcomes. Recent advancements highlight the importance of targeting additional pathways such as mitochondrial dysfunction and neuroinflammation to develop more effective treatments. Personalized medicine, including genetic testing and biomarkers, is proving valuable in stratifying patients and tailoring treatment options. Complementary therapies, such as nutritional interventions like the ketogenic diet and microbiome modulation, also show promise. This review emphasizes the need for a multidisciplinary approach that integrates early diagnosis, targeted treatments, and supportive care to address the multisystemic nature of ALS and improve the quality of life for patients.
Collapse
Affiliation(s)
- Samira Nabakhteh
- Department of Biochemistry, School of Basic Sciences, Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | - Anahita Lotfi
- Department of Food Sciences and Industry, School of Agricultural Sciences and Natural Resources, Islamic Azad University, Khorasgan Branch, Isfahan, Iran
| | - Arman Afsartaha
- Department of Nutrition, Faculty of Medical Sciences and Technologies, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Elaheh Sadat Khodadadi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, 35122, Italy
| | - Siavash Abdolghaderi
- Department of Physical Medicine and Rehabilitation, Iran University of Medical Sciences, Tehran, Iran
| | - Mozhdeh Mohammadpour
- Department of Physical Medicine and Rehabilitation, Iran University of Medical Sciences, Tehran, Iran
| | - Yasaman Shokri
- Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Pouria Kiani
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sajad Ehtiati
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Khakshournia
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Autophagy Research Center, Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Mattson MP. The cyclic metabolic switching theory of intermittent fasting. Nat Metab 2025:10.1038/s42255-025-01254-5. [PMID: 40087409 DOI: 10.1038/s42255-025-01254-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/19/2025] [Indexed: 03/17/2025]
Abstract
Intermittent fasting (IF) and ketogenic diets (KDs) have recently attracted much attention in the scientific literature and in popular culture and follow a longer history of exercise and caloric restriction (CR) research. Whereas IF involves cyclic metabolic switching (CMS) between ketogenic and non-ketogenic states, KDs and CR may not. In this Perspective, I postulate that the beneficial effects of IF result from alternating between activation of adaptive cellular stress response pathways during the fasting period, followed by cell growth and plasticity pathways during the feeding period. Thereby, I establish the cyclic metabolic switching (CMS) theory of IF. The health benefits of IF may go beyond those seen with continuous CR or KDs without CMS owing to the unique interplay between the signalling functions of the ketone β-hydroxybutyrate, mitochondrial adaptations, reciprocal activation of autophagy and mTOR pathways, endocrine and paracrine signalling, gut microbiota, and circadian biology. The CMS theory may have important implications for future basic research, clinical trials, development of pharmacological interventions, and healthy lifestyle practices.
Collapse
Affiliation(s)
- Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
13
|
Shen DM, Byth KF, Bertheloot D, Braams S, Bradley S, Dean D, Dekker C, El-Kattan AF, Franchi L, Glick GD, Ghosh S, Hinniger A, Katz JD, Kitanovic A, Lu X, Olhava EJ, Opipari AW, Sanchez B, Seidel HM, Stunden J, Stutz A, Telling A, Venkatraman S, Winkler DG, Roush WR. Discovery of DFV890, a Potent Sulfonimidamide-Containing NLRP3 Inflammasome Inhibitor. J Med Chem 2025; 68:5529-5550. [PMID: 40036600 DOI: 10.1021/acs.jmedchem.4c02759] [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: 03/06/2025]
Abstract
The discovery of DFV890 ((R)-1), a potent and selective NLRP3 antagonist, is described. Replacement of the sulfonyl urea core from the first-generation NLRP3 antagonist CRID3 with a sulfonimidamide core afforded a novel and potent series of NLRP3 antagonists. The (R)-enantiomers of the sulfonimidamide series were found to be consistently more potent than structurally related sulfonyl ureas. Replacement of the furan unit of CRID3 with a 5-substituted thiazole unit led to DFV890 ((R)-1), which potently inhibited IL-1β production in THP-1 cells and in primary human cells, blocked multiple downstream effectors of NLRP3 activation, and substantially improved PK properties and significantly lowered the predicted human dose compared to that for CRID3. DFV890 ((R)-1) was also effective in an air pouch model of gout.
Collapse
Affiliation(s)
- Dong-Ming Shen
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Kate F Byth
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | | | - Sarah Bradley
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Dennis Dean
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Carien Dekker
- Novartis Biomedical Research, Basel CH-4002, Switzerland
| | | | - Luigi Franchi
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Gary D Glick
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - Shomir Ghosh
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | - Jason D Katz
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | - Xiaokang Lu
- IFM Therapeutics, Ann Arbor, Michigan 48108, United States
| | - Edward J Olhava
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | - Brian Sanchez
- IFM Therapeutics, Ann Arbor, Michigan 48108, United States
| | - H Martin Seidel
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | | | | | - Alissa Telling
- IFM Therapeutics, Ann Arbor, Michigan 48108, United States
| | | | - David G Winkler
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| | - William R Roush
- IFM Therapeutics, Boston, Massachusetts 02116, United States
| |
Collapse
|
14
|
Gregory JM, Smith TJ, Duffus SH, Brooks D, Akbar MN, Huntley MA, Gottlieb JA, LeStourgeon LM, Wilson CS, Beckman JA, Cherrington AD. A one-week reduced-carbohydrate diet to mitigate iatrogenic peripheral hyperinsulinemia does not improve insulin sensitivity or endothelial function in a randomized, crossover trial in patients with type 1 diabetes. Cardiovasc Diabetol 2025; 24:107. [PMID: 40045281 PMCID: PMC11884211 DOI: 10.1186/s12933-025-02658-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Iatrogenic peripheral hyperinsulinemia, resulting from peripheral insulin administration in type 1 diabetes, may increase insulin resistance and impair endothelial function. We hypothesized that lowering iatrogenic peripheral hyperinsulinemia via a one-week, reduced-carbohydrate diet (RCD) would improve insulin sensitivity and endothelial function compared with an isocaloric standard carbohydrate diet (SCD). METHODS In this randomized, single-blinded, crossover trial, we studied 12 adults with type 1 diabetes. Participants completed both a one-week RCD and a one-week SCD, separated by a three-week washout. After each intervention, we measured insulin sensitivity using a hyperinsulinemic-euglycemic clamp and assessed endothelial function via brachial-artery flow-mediated vasodilation (FMD). RESULTS The RCD reduced total daily insulin doses by 16% compared with the SCD. Despite this reduction, insulin sensitivity did not improve (median glucose infusion rates: RCD 8.1 mg/kg FFM/min [IQR 6.7-10.1] vs. SCD 8.6 mg/kg FFM/min [7.0-11.0], p = 0.47). Similarly, endothelial function did not differ significantly (FMD after RCD 7.50% [3.25-15.5] vs. SCD 9.81% [4.96-14.3], p = 0.91). Although higher insulin doses correlated with lower insulin sensitivity under both conditions, lowering insulin dose through the RCD alone did not yield measurable improvements. CONCLUSIONS Although a one-week RCD significantly lowered insulin requirements, it failed to enhance insulin sensitivity or endothelial function in adults with type 1 diabetes. These findings underscore the complex and dynamic relationship between insulin exposure and cardiometabolic health. Similar basal overnight insulin delivery may have masked potential benefits by the time of testing, highlighting the need for further studies to refine strategies aimed at mitigating hyperinsulinemia's adverse effects. TRIAL REGISTRATION ClinicalTrials.gov NCT04118374.
Collapse
Affiliation(s)
- Justin M Gregory
- Ian Burr Division of Pediatric Endocrinology and Diabetes, Vanderbilt University School of Medicine, 1500 21st Avenue South, Suite 1514, Nashville, TN, 37212-3157, USA.
| | - T Jordan Smith
- Ian Burr Division of Pediatric Endocrinology and Diabetes, Vanderbilt University School of Medicine, 1500 21st Avenue South, Suite 1514, Nashville, TN, 37212-3157, USA
| | - Sara H Duffus
- Ian Burr Division of Pediatric Endocrinology and Diabetes, Vanderbilt University School of Medicine, 1500 21st Avenue South, Suite 1514, Nashville, TN, 37212-3157, USA
- Division of Pediatric Endocrinology, University of North Carolina, 127 Medical School Wing E, CB# 7039, Chapel Hill, NC, 27599-7039, USA
| | - David Brooks
- Mildred Stahlman Division of Neonatology, Vanderbilt University School of Medicine, 1161 21st Ave S A0126, Nashville, TN, 37232, USA
- Department of Pediatrics, Creighton University, 2412 Cuming St #103, Omaha, NE, 68131, USA
| | - M Naweed Akbar
- Ian Burr Division of Pediatric Endocrinology and Diabetes, Vanderbilt University School of Medicine, 1500 21st Avenue South, Suite 1514, Nashville, TN, 37212-3157, USA
| | - Margaret-Anne Huntley
- Ian Burr Division of Pediatric Endocrinology and Diabetes, Vanderbilt University School of Medicine, 1500 21st Avenue South, Suite 1514, Nashville, TN, 37212-3157, USA
| | - JoAnn A Gottlieb
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University School of Medicine, 2525 West End Ave., Nashville, TN, 37203-8820, USA
| | - Lauren M LeStourgeon
- Department of Internal Medicine, Vanderbilt University School of Medicine, 1161 21st Ave S, Nashville, TN, 37232, USA
| | - Christopher S Wilson
- Ian Burr Division of Pediatric Endocrinology and Diabetes, Vanderbilt University School of Medicine, 1500 21st Avenue South, Suite 1514, Nashville, TN, 37212-3157, USA
| | - Joshua A Beckman
- Division of Vascular Medicine, University of Texas Southwestern, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2301 Vanderbilt Place, Nashville, TN, 37240, USA
| |
Collapse
|
15
|
Schaible P, Henschel J, Erny D. How the gut microbiota impacts neurodegenerative diseases by modulating CNS immune cells. J Neuroinflammation 2025; 22:60. [PMID: 40033338 DOI: 10.1186/s12974-025-03371-0] [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: 01/08/2025] [Accepted: 02/06/2025] [Indexed: 03/05/2025] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide. Amyloid-β (Aβ) accumulation and neurofibrillary tangles are two key histological features resulting in progressive and irreversible neuronal loss and cognitive decline. The macrophages of the central nervous system (CNS) belong to the innate immune system and comprise parenchymal microglia and CNS-associated macrophages (CAMs) at the CNS interfaces (leptomeninges, perivascular space and choroid plexus). Microglia and CAMs have received attention as they may play a key role in disease onset and progression e. g., by clearing amyloid beta (Aβ) through phagocytosis. Genome-wide association studies (GWAS) have revealed that human microglia and CAMs express numerous risk genes for AD, further highlighting their potentially critical role in AD pathogenesis. Microglia and CAMs are tightly controlled by environmental factors, such as the host microbiota. Notably, it was further reported that the composition of the gut microbiota differed between AD patients and healthy individuals. Hence, emerging studies have analyzed the impact of gut bacteria in different preclinical mouse models for AD as well as in clinical studies, potentially enabling promising new therapeutic options.
Collapse
Affiliation(s)
- Philipp Schaible
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Julia Henschel
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Daniel Erny
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany.
| |
Collapse
|
16
|
Bouck T, Monteleone J, Duffy J, Ainslie PN, Little JP, Thomas KN, Gibbons TD, Islam H. Changes in plasma cytokines following a 60-h fast are not influenced by the addition of exercise despite elevated ketones in healthy young adults. Physiol Rep 2025; 13:e70294. [PMID: 40129260 PMCID: PMC11933719 DOI: 10.14814/phy2.70294] [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: 12/24/2024] [Revised: 02/24/2025] [Accepted: 03/18/2025] [Indexed: 03/26/2025] Open
Abstract
Immunometabolic processes maintain physiological homeostasis and are implicated in various chronic diseases. Fasting and exercise independently alter metabolic and immunological processes; their combination could provide insights into immunometabolic interactions. Using a randomized crossover design, 15 healthy adults (six females, nine males, 26.5 ± 4.3 years) fasted for 60 h with and without the addition of a 3 h cycling bout (65%-80% VO2 peak). Fasting alone (FAST) and with exercise (FEX) reduced plasma glucose, insulin, respiratory exchange ratio, and increased β-hydroxybutyrate (BHB; all p < 0.01). FEX elicited more rapid changes in glucose and BHB and higher BHB concentrations at 60 h (all p < 0.01). Both conditions decreased circulating TNF-⍺ concentrations and increased IL-10 (p < 0.01), although the increase in IL-10 appeared to be driven by the FEX condition (p = 0.03). IL-6 concentrations tended to increase in both conditions (p = 0.1). Total white blood cell count remained unchanged after 60 h in both conditions, with only modest changes in some leukocyte subpopulations. Collectively, the observed changes in circulating cytokine concentrations support an overall anti-inflammatory effect of prolonged fasting, while the maintenance of leukocyte concentrations suggests immune function is not compromised. Despite greater metabolic strain, the addition of prolonged exercise did not appear to augment changes in systemic cytokines and leukocytes.
Collapse
Affiliation(s)
- Tori Bouck
- School of Health and Exercise Sciences, The University of British Columbia OkanaganKelownaBritish ColumbiaCanada
| | - Justin Monteleone
- School of Health and Exercise Sciences, The University of British Columbia OkanaganKelownaBritish ColumbiaCanada
| | - Jennifer Duffy
- School of Health and Exercise Sciences, The University of British Columbia OkanaganKelownaBritish ColumbiaCanada
| | - Philip N. Ainslie
- School of Health and Exercise Sciences, The University of British Columbia OkanaganKelownaBritish ColumbiaCanada
- Centre for Heart, Lung and Vascular HealthSchool of Health and Exercise Sciences, The University of British Columbia OkanaganKelownaBritish ColumbiaCanada
| | - Jonathan P. Little
- School of Health and Exercise Sciences, The University of British Columbia OkanaganKelownaBritish ColumbiaCanada
- Centre for Chronic Disease Prevention and Management, Faculty of MedicineThe University of British Columbia OkanaganKelownaBritish ColumbiaCanada
| | - Kate N. Thomas
- Department of Surgical SciencesDunedin School of Medicine, University of OtagoDunedinNew Zealand
| | - Travis D. Gibbons
- Department of Biological SciencesNorthern Arizona UniversityFlagstaffArizonaUSA
| | - Hashim Islam
- School of Health and Exercise Sciences, The University of British Columbia OkanaganKelownaBritish ColumbiaCanada
- Centre for Chronic Disease Prevention and Management, Faculty of MedicineThe University of British Columbia OkanaganKelownaBritish ColumbiaCanada
| |
Collapse
|
17
|
Wang H, Ma L, Su W, Liu Y, Xie N, Liu J. NLRP3 inflammasome in health and disease (Review). Int J Mol Med 2025; 55:48. [PMID: 39930811 PMCID: PMC11781521 DOI: 10.3892/ijmm.2025.5489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/16/2024] [Indexed: 02/13/2025] Open
Abstract
Activation of inflammasomes is the activation of inflammation‑related caspase mediated by the assembly signal of multi‑protein complex and the maturity of inflammatory factors, such as IL‑1β and IL‑18. Among them, the Nod‑like receptor family pyrin domain containing 3 (NLRP3) inflammasome is the most thoroughly studied type of inflammatory corpuscle at present, which is involved in the occurrence and development of numerous human diseases. Therefore, targeting the NLRP3 inflammasome has become the focus of drug development for related diseases. In this paper, the research progress of the NLRP3 inflammasome in recent years is summarized, including the activation and regulation of NLRP3 and its association with diseases. A deep understanding of the regulatory mechanism of NLRP3 will be helpful to the discovery of new drug targets and the development of therapeutic drugs.
Collapse
Affiliation(s)
- Haoran Wang
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Li Ma
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Weiran Su
- Department of Internal Medicine, Jiading District Central Hospital, Shanghai 201800, P.R. China
| | - Yangruoyu Liu
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Ning Xie
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Jun Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| |
Collapse
|
18
|
Kim HN, Lee JH, Boscardin J, Newman JC. Plasma β-hydroxybutyrate concentration, genetic risk, and the incidence of Alzheimer's disease: A prospective study of 261,933 participants. Clin Nutr 2025; 46:1-9. [PMID: 39854811 DOI: 10.1016/j.clnu.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/11/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND We investigated whether plasma β-hydroxybutyrate levels, a genetic risk score for Alzheimer's disease, and their interaction are associated with incident Alzheimer's disease. METHODS Using data from the UK Biobank-a population-based cohort study of adults aged 40-69 years, we assessed associations between baseline plasma β-hydroxybutyrate level, genetic risk score for Alzheimer's disease, and incident Alzheimer's disease. Incident Alzheimer's disease data were collected through linked data from hospital admissions and death registries. RESULTS In total, 261,933 adults were included, 1978 of whom developed incident Alzheimer's disease. Plasma β-hydroxybutyrate concentrations were not independently associated with Alzheimer's disease incidence after adjusting for covariates, whereas a higher genetic predisposition was linked to increased Alzheimer's disease incidence. Interactions were observed between plasma β-hydroxybutyrate concentrations and genetic risk for Alzheimer's disease on Alzheimer's disease incidence (P < 0.001). CONCLUSIONS Further studies are warranted to elucidate the impact of plasma β-hydroxybutyrate status on Alzheimer's disease incidence.
Collapse
Affiliation(s)
- Ha-Na Kim
- Department of Family Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, 93, Jungbu-daero, Paldal-gu, Suwon-si, Gyeonggi-do, 16247, Republic of Korea.
| | - Ji Hyun Lee
- Department of Dermatology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| | - John Boscardin
- Division of Geriatrics, Department of Medicine, School of Medicine, University of California San Francisco, 490 Illinois Street, Floor 8, San Francisco, CA, 94143, USA.
| | - John C Newman
- Division of Geriatrics, Department of Medicine, School of Medicine, University of California San Francisco, 490 Illinois Street, Floor 8, San Francisco, CA, 94143, USA; Buck Institute for Research on Aging, Novato, CA, 94945, USA.
| |
Collapse
|
19
|
Wang R, Liao Y, Deng Y, Shuang R. Unraveling the Health Benefits and Mechanisms of Time-Restricted Feeding: Beyond Caloric Restriction. Nutr Rev 2025; 83:e1209-e1224. [PMID: 38954563 DOI: 10.1093/nutrit/nuae074] [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] [Indexed: 07/04/2024] Open
Abstract
Time-restricted feeding (TRF) is a lifestyle intervention that aims to maintain a consistent daily cycle of feeding and fasting to support robust circadian rhythms. Recently, it has gained scientific, medical, and public attention due to its potential to enhance body composition, extend lifespan, and improve overall health, as well as induce autophagy and alleviate symptoms of diseases like cardiovascular diseases, type 2 diabetes, neurodegenerative diseases, cancer, and ischemic injury. However, there is still considerable debate on the primary factors that contribute to the health benefits of TRF. Despite not imposing strict limitations on calorie intake, TRF consistently led to reductions in calorie intake. Therefore, while some studies suggest that the health benefits of TRF are primarily due to caloric restriction (CR), others argue that the key advantages of TRF arise not only from CR but also from factors like the duration of fasting, the timing of the feeding period, and alignment with circadian rhythms. To elucidate the roles and mechanisms of TRF beyond CR, this review incorporates TRF studies that did not use CR, as well as TRF studies with equivalent energy intake to CR, which addresses the previous lack of comprehensive research on TRF without CR and provides a framework for future research directions.
Collapse
Affiliation(s)
- Ruhan Wang
- Department of Nutrition Hygiene and Toxicology, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 43000, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 43000, China
| | - Yan Deng
- Department of Nutrition Hygiene and Toxicology, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 43000, China
| | - Rong Shuang
- Department of Nutrition Hygiene and Toxicology, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 43000, China
| |
Collapse
|
20
|
Mullin SM, Kelly AJ, Ní Chathail MB, Norris S, Shannon CE, Roche HM. Macronutrient Modulation in Metabolic Dysfunction-Associated Steatotic Liver Disease-the Molecular Role of Fatty Acids compared with Sugars in Human Metabolism and Disease Progression. Adv Nutr 2025; 16:100375. [PMID: 39842721 PMCID: PMC11849631 DOI: 10.1016/j.advnut.2025.100375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/23/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a significant public health concern, with its progression to metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis leading to severe outcomes including cirrhosis, hepatocellular carcinoma, and liver failure. Whereas obesity and excess energy intake are well-established contributors to the development and progression of MASLD, the distinct role of specific macronutrients is less clear. This review examines the mechanistic pathways through which dietary fatty acids and sugars contribute to the development of hepatic inflammation and fibrosis, offering a nuanced understanding of their respective roles in MASLD progression. In terms of addressing potential therapeutic options, human intervention studies that investigate whether modifying the intake of dietary fats and carbohydrates affects MASLD progression are reviewed. By integrating this evidence, this review seeks to bridge the gap in the understanding between the mechanisms of macronutrient-driven MASLD progression and the effect of altering the intake of these nutrients in the clinical setting and presents a foundation for future research into targeted dietary strategies for the treatment of the disease.
Collapse
Affiliation(s)
- Sinéad M Mullin
- School of Public Health, Physiotherapy and Sport Science, and Institute of Food and Health, University College Dublin, Belfield, Dublin, Ireland; Nutrigenomics Research Group, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Aidan J Kelly
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Méabh B Ní Chathail
- School of Public Health, Physiotherapy and Sport Science, and Institute of Food and Health, University College Dublin, Belfield, Dublin, Ireland; Nutrigenomics Research Group, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Suzanne Norris
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Christopher E Shannon
- Nutrigenomics Research Group, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland; School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Helen M Roche
- School of Public Health, Physiotherapy and Sport Science, and Institute of Food and Health, University College Dublin, Belfield, Dublin, Ireland; Nutrigenomics Research Group, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland; Institute for Global Food Security, Queen's University Belfast, Northern Ireland.
| |
Collapse
|
21
|
Krejčová G, Novotná D, Bajgar A. Ketogenesis nutritionally supports brain during bacterial infection in Drosophila. Brain Behav Immun 2025; 125:280-291. [PMID: 39824470 DOI: 10.1016/j.bbi.2025.01.008] [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: 08/12/2024] [Revised: 12/19/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025] Open
Abstract
Mounting an immune response is a nutritionally demanding process that requires the systemic redistribution of energy stores towards the immune system. This is facilitated by cytokine-induced insulin resistance, which simultaneously promotes the mobilization of lipids and carbohydrates while limiting their consumption in immune-unrelated processes, such as development, growth, and reproduction. However, this adaptation also restricts the availability of nutrients to vital organs, which must then be sustained by alternative fuels. Here, we employed an experimental model of severe bacterial infection in Drosophila melanogaster to investigate whether ketogenesis may represent a metabolic adaptation for overcoming periods of nutritional scarcity during the immune response. We found that the immune response to severe bacterial infection is accompained by increased ketogenesis in the fat body and macrophages, leading to elevated levels of β-hydroxybutyrate in circulation. Although this metabolic adaptation is essential for survival during infection, it is not required for the elimination of the pathogen itself. Instead, ketone bodies predominately serve as an energy source for the brain neurons during this period of nutrient scarcity.
Collapse
Affiliation(s)
- Gabriela Krejčová
- University of South Bohemia, Faculty of Science, Department of Molecular Biology and Genetics, Ceske Budejovice, Czech Republic
| | - Diana Novotná
- University of South Bohemia, Faculty of Science, Department of Molecular Biology and Genetics, Ceske Budejovice, Czech Republic
| | - Adam Bajgar
- University of South Bohemia, Faculty of Science, Department of Molecular Biology and Genetics, Ceske Budejovice, Czech Republic.
| |
Collapse
|
22
|
Wong A, Sun Q, Latif II, Karwi QG. Macrophage energy metabolism in cardiometabolic disease. Mol Cell Biochem 2025; 480:1763-1783. [PMID: 39198360 PMCID: PMC11842501 DOI: 10.1007/s11010-024-05099-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
In a rapidly expanding body of literature, the major role of energy metabolism in determining the response and polarization status of macrophages has been examined, and it is currently a very active area of research. The metabolic flux through different metabolic pathways in the macrophage is interconnected and complex and could influence the polarization of macrophages. Earlier studies suggested glucose flux through cytosolic glycolysis is a prerequisite to trigger the pro-inflammatory phenotypes of macrophages while proposing that fatty acid oxidation is essential to support anti-inflammatory responses by macrophages. However, recent studies have shown that this understanding is oversimplified and that the metabolic control of macrophage polarization is highly complex and not fully defined yet. In this review, we systematically reviewed and summarized the literature regarding the role of energy metabolism in controlling macrophage activity and how that might be altered in cardiometabolic diseases, namely heart failure, obesity, and diabetes. We critically appraised the experimental studies and methodologies in the published studies. We also highlighted the challenging concepts in macrophage metabolism and identified several research questions yet to be addressed in future investigations.
Collapse
Affiliation(s)
- Angela Wong
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Qiuyu Sun
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Ismail I Latif
- Department of Microbiology, College of Medicine, University of Diyala, Baqubaa, Diyala, Iraq
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1B 3V6, Canada.
| |
Collapse
|
23
|
Freyberg Z, Andreazza AC, McClung CA, Phillips ML. Linking Mitochondrial Dysfunction, Neurotransmitter, and Neural Network Abnormalities and Mania: Elucidating Neurobiological Mechanisms of the Therapeutic Effect of the Ketogenic Diet in Bipolar Disorder. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2025; 10:267-277. [PMID: 39053576 PMCID: PMC11754533 DOI: 10.1016/j.bpsc.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/25/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
There is growing interest in the ketogenic diet as a treatment for bipolar disorder (BD), and there are promising anecdotal and small case study reports of efficacy. However, the neurobiological mechanisms by which diet-induced ketosis might ameliorate BD symptoms remain to be determined, particularly in manic and hypomanic states-defining features of BD. Identifying these mechanisms will provide new markers to guide personalized interventions and provide targets for novel treatment developments for individuals with BD. In this critical review, we describe recent findings highlighting 2 types of neurobiological abnormalities in BD: 1) mitochondrial dysfunction and 2) neurotransmitter and neural network functional abnormalities. We link these abnormalities to mania/hypomania and depression in BD and then describe the biological underpinnings by which the ketogenic diet may have a beneficial effect in individuals with BD. We end the review by describing approaches that can be employed in future studies to elucidate the neurobiology that underlies the therapeutic effect of the ketogenic diet in BD. Doing this may provide marker predictors to identify individuals who will respond well to the ketogenic diet, as well as offer neural targets for novel treatment developments for BD.
Collapse
Affiliation(s)
- Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Ana C Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Colleen A McClung
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mary L Phillips
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
24
|
Balde A, Benjakul S, Nazeer RA. A review on NLRP3 inflammasome modulation by animal venom proteins/peptides: mechanisms and therapeutic insights. Inflammopharmacology 2025; 33:1013-1031. [PMID: 39934538 DOI: 10.1007/s10787-025-01656-7] [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: 12/06/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025]
Abstract
The venom peptides from terrestrial as well as aquatic species have demonstrated potential in regulating the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a sophisticated assemblage present in immune cells responsible for detecting and responding to external mediators. The NLRP3 inflammasome plays a role in several pathological conditions such as type 2 diabetes, hyperglycemia, Alzheimer's disease, obesity, autoimmune disorders, and cardiovascular disorders. Venom peptides derived from animal venoms have been discovered to selectively induce certain signalling pathways, such as the NLRP3 inflammasome, mitogen-activated protein kinase (MAPK), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Experimental evidence has demonstrated that venom peptides can regulate the expression and activation of the NLRP3 inflammasome, resulting in the secretion of pro-inflammatory cytokines including interleukin (IL)-1β and IL-18. Furthermore, these peptides have been discovered to impede the activation of the NLRP3 inflammasome, therefore diminishing inflammation and tissue injury. The functional properties of venom proteins and peptides obtained from snakes, bees, wasps, and scorpions have been thoroughly investigated, specifically targeting the NLRP3 inflammasome pathway, venom proteins and peptides have shown promise as therapeutic agents for the treatment of certain inflammatory disorders. This review discusses the pathophysiology of NLRP3 inflammasome in the onset of various diseases, role of venom as therapeutics. Further, various venom components and their role in the modulation of NLRP3 inflammasome are discoursed. A substantial number of venomous animals and their toxins are yet unexplored, and to comprehensively grasp the mechanisms of action of them and their potential as therapeutic agents, additional research is required which can lead to the development of novel therapeutics.
Collapse
Affiliation(s)
- Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603 203, India
| | - Soottawat Benjakul
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro Industry, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand
- Department of Food and Nutrition, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603 203, India.
| |
Collapse
|
25
|
Vieira AB, Cavanaugh SM, Ciambarella BT, Machado MV. Sodium-glucose co-transporter 2 inhibitors: a pleiotropic drug in humans with promising results in cats. Front Vet Sci 2025; 12:1480977. [PMID: 40093620 PMCID: PMC11906673 DOI: 10.3389/fvets.2025.1480977] [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: 08/14/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Diabetes mellitus is a common metabolic disease in humans and cats. Cats share several features of human type-2 diabetes and can be considered an animal model for this disease. In the last decade, sodium-glucose transporter 2 inhibitors (SGLT2i) have been used successfully as a class of hypoglycemic drug that inhibits the reabsorption of glucose from the renal proximal tubules, consequently managing hyperglycemia through glycosuria. Furthermore, SGLT2i have been shown to have cardiac, renal, and other protective effects in diabetic humans acting as a pleiotropic drug. Currently, at least six SGLT2i are approved by the Food and Drug Administration (FDA) for use in humans with type-2 diabetes, and recently, two drugs were approved for use in diabetic cats. This narrative review focuses on the use of SGLT2i to treat diabetes mellitus in humans and cats. We summarize the human data that support the use of SGLT2i in controlling type-2 diabetes and protecting against cardiovascular and renal damage. We also review the available literature regarding other benefits of these drugs in humans as well as the effects of SGLT2i in cats. Adverse effects related to the use of these hypoglycemic drugs are also discussed.
Collapse
Affiliation(s)
- Aline B. Vieira
- Biomedical Sciences Department, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Sarah M. Cavanaugh
- Department of Clinical Sciences, Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
| | - Bianca T. Ciambarella
- Laboratory of Ultrastructure and Tissue Biology, Anatomy Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcus V. Machado
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
| |
Collapse
|
26
|
Džidić Krivić A, Begagić E, Hadžić S, Bećirović A, Bećirović E, Hibić H, Tandir Lihić L, Kadić Vukas S, Bečulić H, Kasapović T, Pojskić M. Unveiling the Important Role of Gut Microbiota and Diet in Multiple Sclerosis. Brain Sci 2025; 15:253. [PMID: 40149775 PMCID: PMC11939953 DOI: 10.3390/brainsci15030253] [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: 12/27/2024] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS), characterized by neurodegeneration, axonal damage, demyelination, and inflammation. Recently, gut dysbiosis has been linked to MS and other autoimmune conditions. Namely, gut microbiota has a vital role in regulating immune function by influencing immune cell development, cytokine production, and intestinal barrier integrity. While balanced microbiota fosters immune tolerance, dysbiosis disrupts immune regulation, damages intestinal permeability, and heightens the risk of autoimmune diseases. The critical factor in shaping the gut microbiota and modulating immune response is diet. Research shows that high-fat diets rich in saturated fats are associated with disease progression. Conversely, diets rich in fruits, yogurt, and legumes may lower the risk of MS onset and progression. Specific dietary interventions, such as the Mediterranean diet (MD) and ketogenic diet, have shown potential to reduce inflammation, support neuroprotection, and promote CNS repair. Probiotics, by restoring microbial balance, may also help mitigate immune dysfunction noted in MS. Personalized dietary strategies targeting the gut microbiota hold promise for managing MS by modulating immune responses and slowing disease progression. Optimizing nutrient intake and adopting anti-inflammatory diets could improve disease control and quality of life. Understanding gut-immune interactions is essential for developing tailored nutritional therapies for MS patients.
Collapse
Affiliation(s)
- Amina Džidić Krivić
- Department of Neurology, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina; (A.D.K.); (L.T.L.)
- Department of Physiology, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina
| | - Emir Begagić
- Department of Neurosurgery, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina; (E.B.)
- Department of Doctoral Studies, School of Medicine, University of Tuzla, 75000 Tuzla, Bosnia and Herzegovina
| | - Semir Hadžić
- Internal Medicine Clinic, University Clinical Center of Tuzla, Ulica prof. dr. Ibre Pašića, 75000 Tuzla, Bosnia and Herzegovina (E.B.)
- Department of Physiology, School of Medicine, University of Tuzla, Univerzitetska 1, 75000 Tuzla, Bosnia and Herzegovina
| | - Amir Bećirović
- Internal Medicine Clinic, University Clinical Center of Tuzla, Ulica prof. dr. Ibre Pašića, 75000 Tuzla, Bosnia and Herzegovina (E.B.)
| | - Emir Bećirović
- Internal Medicine Clinic, University Clinical Center of Tuzla, Ulica prof. dr. Ibre Pašića, 75000 Tuzla, Bosnia and Herzegovina (E.B.)
| | - Harisa Hibić
- Department of Maxillofacial Surgery, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina
| | - Lejla Tandir Lihić
- Department of Neurology, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina; (A.D.K.); (L.T.L.)
- Department of Neurology, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina
| | - Samra Kadić Vukas
- Department of Neurology, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina; (A.D.K.); (L.T.L.)
- Department of Neurology, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina
| | - Hakija Bečulić
- Department of Neurosurgery, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina; (E.B.)
- Department of Anatomy, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina
| | - Tarik Kasapović
- Internal Medicine Clinic, University Clinical Center of Tuzla, Ulica prof. dr. Ibre Pašića, 75000 Tuzla, Bosnia and Herzegovina (E.B.)
| | - Mirza Pojskić
- Department of Neurosurgery, University Hospital Marburg, Baldingerstr., 35033 Marburg, Germany
| |
Collapse
|
27
|
Fastiggi VA, Mank MM, Caporizzo MA, Poynter ME. Beta-Hydroxybutyrate Inhibits Bronchial Smooth Muscle Contraction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639075. [PMID: 40060651 PMCID: PMC11888348 DOI: 10.1101/2025.02.24.639075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
Asthma is a chronic respiratory condition characterized by airway inflammation, remodeling, and hyperresponsiveness to triggers causing airway constriction. Bronchial smooth muscle plays a critical role by narrowing airways, leading to obstruction and breathing difficulties, often exacerbated by mast cell infiltration and histamine release. Whereas current treatments, including bronchodilators, corticosteroids, and biologics provide effective management for most patients, alternative therapies are needed for difficult-to-treat asthma. Recent research highlights the potential of therapeutic ketosis, achieved through dietary interventions or supplementation with exogenous ketones, to reduce airway hyperresponsiveness and inflammation. Ketone bodies, known for providing energy during carbohydrate scarcity, also influence asthma by activating cell-surface receptors and transporters. In vivo, interventions like weight loss and caloric restriction increase ketone body levels, correlating with improved asthma symptoms, reduced oxidative stress, and inflammation. These effects suggest ketone bodies, particularly β-hydroxybutyrate, may play a therapeutic role in mitigating bronchoconstriction and smooth muscle contraction in asthma. We utilize human bronchial smooth muscle cells (in vitro) and mouse precision-cut lung slices (PCLS) (ex vivo) to assess the effects of BHB on histamine-induced bronchoconstriction. Brightfield microscopy showed that BHB reduces contraction in human bronchial smooth muscle cells, an effect involving free fatty acid receptor 3 (FFAR3) activation. Light microscopy of PCLS revealed that BHB inhibits airway narrowing and cellular extrusion, demonstrating its ability to mitigate bronchoconstriction by suppressing smooth muscle contraction. These results implicate bronchial smooth muscle as a cellular target of therapeutic ketosis, an important contributor to the beneficial effects of BHB in preclinical models of asthma.
Collapse
Affiliation(s)
- V Amanda Fastiggi
- Department of Medicine, Division of Pulmonary Disease and Critical Care, University of Vermont, and The Vermont Lung Center, Burlington, VT, 05405, USA
- Cellular, Molecular, and Biomedical Sciences Doctoral Program, University of Vermont, Burlington, VT, 05405, USA
| | - Madeleine M Mank
- Department of Medicine, Division of Pulmonary Disease and Critical Care, University of Vermont, and The Vermont Lung Center, Burlington, VT, 05405, USA
| | - Matthew A Caporizzo
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, 05405, USA
| | - Matthew E Poynter
- Department of Medicine, Division of Pulmonary Disease and Critical Care, University of Vermont, and The Vermont Lung Center, Burlington, VT, 05405, USA
| |
Collapse
|
28
|
Cote AL, Munger CJ, Ringel AE. Emerging insights into the impact of systemic metabolic changes on tumor-immune interactions. Cell Rep 2025; 44:115234. [PMID: 39862435 DOI: 10.1016/j.celrep.2025.115234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/24/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Tumors are inherently embedded in systemic physiology, which contributes metabolites, signaling molecules, and immune cells to the tumor microenvironment. As a result, any systemic change to host metabolism can impact tumor progression and response to therapy. In this review, we explore how factors that affect metabolic health, such as diet, obesity, and exercise, influence the interplay between cancer and immune cells that reside within tumors. We also examine how metabolic diseases influence cancer progression, metastasis, and treatment. Finally, we consider how metabolic interventions can be deployed to improve immunotherapy. The overall goal is to highlight how metabolic heterogeneity in the human population shapes the immune response to cancer.
Collapse
Affiliation(s)
- Andrea L Cote
- Ragon Institute of Mass General, MIT, and Harvard, 600 Main Street, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, 31 Ames Street, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Chad J Munger
- Ragon Institute of Mass General, MIT, and Harvard, 600 Main Street, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, 31 Ames Street, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Alison E Ringel
- Ragon Institute of Mass General, MIT, and Harvard, 600 Main Street, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, 31 Ames Street, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA.
| |
Collapse
|
29
|
Qiao M, Ni J, Qing H, Qiu Y, Quan Z. Role of Peripheral NLRP3 Inflammasome in Cognitive Impairments: Insights of Non-central Factors. Mol Neurobiol 2025:10.1007/s12035-025-04779-8. [PMID: 40000575 DOI: 10.1007/s12035-025-04779-8] [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: 11/13/2024] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
Cognitive impairments are common clinical manifestation of Alzheimer's disease, vascular dementia, type 2 diabetes mellitus, and autoimmune diseases. Emerging evidence has suggested a strong correlation between peripheral chronic inflammation and cognitive impairments. For example, nearly 40% of individuals with inflammatory bowel disease also suffer from cognitive impairments. In this condition, NLRP3 inflammasome (NLRP3-I) generating pro-inflammatory cytokines like IL-1β serves as a significant effector, and its persistence exerts adverse effects to both periphery and the brain. Moreover, investigations on serum biomarkers of mild cognitive impairments have shown NLRP3-I components' upregulation, suggesting the involvement of peripheral inflammasome pathway in this disorder. Here, we systematically reviewed the current knowledge of NLRP3-I in inflammatory disease to uncover its potential role in bridging peripheral chronic inflammation and cognitive impairments. This review summarizes the molecular features and ignition process of NLRP3-I in inflammatory response. Meanwhile, various effects of NLRP3-I involved in peripheral inflammation-associated disease are also reviewed, especially its chronic disturbances to brain homeostasis and cognitive function through routes including gut-brain, liver-brain, and kidney-brain axes. In addition, current promising compounds and their targets relative to NLRP3-I are discussed in the context of cognitive impairments. Through the detailed investigation, this review highlights the critical role of peripheral NLRP3-I in the pathogenesis of cognitive disorders, and offers novel perspectives for developing effective therapeutic interventions for diseases associated with cognitive impairments. The present review outlines the current knowledge on the ignition of NLRP3-I in inflammatory disease and more importantly, emphasizes the role of peripheral NLRP3-I as a causal pathway in the development of cognitive disorders. Although major efforts to restrain cognitive decline are mainly focused on the central nervous system, it has become clear that disturbances from peripheral immune are closely associated with the dysfunctional brain. Therefore, attenuation of these inflammatory changes through inhibiting the NLRP3-I pathway in early inflammatory disease may reduce future risk of cognitive impairments, and in the meantime, considerations on such pathogenesis for combined drug therapy will be required in the clinical evaluation of cognitive disorders.
Collapse
Affiliation(s)
- Mengfan Qiao
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
- Department of Biology, Shenzhen MSU-BIT University, Shenzhen, 518172, China
| | - Yunjie Qiu
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
30
|
Stephens EB, Senadheera C, Roa-Diaz S, Peralta S, Alexander L, Silverman-Martin W, Yukawa M, Morris J, Johnson JB, Newman JC, Stubbs BJ. A Randomized Open-Label, Observational Study of the Novel Ketone Ester, Bis Octanoyl (R)-1,3-Butanediol, and Its Acute Effect on ß-Hydroxybutyrate and Glucose Concentrations in Healthy Older Adults. J Nutr Gerontol Geriatr 2025:1-20. [PMID: 39985761 DOI: 10.1080/21551197.2025.2466163] [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: 02/24/2025]
Abstract
Bis-octanoyl-(R)-1,3-butanediol (BO-BD) is a novel ketone ester (KE) ingredient which increases blood beta-hydroxybutyrate (BHB) concentration rapidly after ingestion. KE is hypothesized to improve function in older adults. Whilst many studies have investigated KE in young adults, they have not been studied in healthy older adults (HOA), for whom age-related differences in metabolism may alter the effects. This randomized, observational, open-label study in HOA (n = 30, 50% male, age = 76.5y) aimed to elucidate tolerance, blood BHB and glucose concentrations for 4h following consumption of either 12.5 or 25 g of BO-BD formulated in ready-to-drink beverage (n = 30), and re-constituted powder (n = 21) with a meal. All study interventions were well tolerated, and increased blood BHB, inducing nutritional ketosis (≥0.5 mM) until the end of the study. Peak BHB concentration (Cmax) and incremental area under the curve (iAUC) were significantly greater with 25 vs 12.5 g of BO-BD in both formulations. There were no significant differences in Cmax or iAUC between formulations. Blood glucose increased in all conditions following the meal, with no consistent significant differences between conditions. These results demonstrate that both powder and beverage formulations of the KE, BO-BD, induce ketosis in HOA adults, facilitating future research on functional effects of KE in aging.
Collapse
Affiliation(s)
| | | | | | - Sawyer Peralta
- Buck Institute for Research on Aging, Novato, California, USA
| | - Laura Alexander
- Buck Institute for Research on Aging, Novato, California, USA
| | | | - Michi Yukawa
- Veteran's Affairs Medical Center, San Francisco, California, USA
| | - Jennifer Morris
- Veteran's Affairs Medical Center, San Francisco, California, USA
| | | | - John C Newman
- Buck Institute for Research on Aging, Novato, California, USA
- Division of Geriatrics, University of California, San Francisco, California, USA
| | | |
Collapse
|
31
|
Fastiggi VA, Mank MM, Poynter ME. Beta-Hydroxybutyrate Attenuates Bronchial Smooth Muscle Pro-Inflammatory Cytokine Production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.639048. [PMID: 40027689 PMCID: PMC11870512 DOI: 10.1101/2025.02.19.639048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Asthma is a common airway condition causing breathing difficulties due to reversible airflow obstruction. It often affects obese individuals, with symptoms triggered by environmental factors that induce immune responses, leading to inflammation and bronchoconstriction. Bronchial smooth muscle (BSM) plays a central role in airway narrowing, driven by type 2 immune responses involving cytokines like IL-4, IL-5, and IL-13, along with leukocytes including eosinophils and type 2 T-helper cells. These responses cause structural changes such as fibrosis and airway thickening, while BSM cells worsen asthma by releasing pro-inflammatory cytokines in response to allergens, microbial signals, or inflammatory cytokines from other cells. While current treatments manage asthma in most patients, alternative therapies are needed for difficult-to-treat cases, particularly prevalent in obese, allergic individuals. Emerging research suggests that therapeutic ketosis, induced by dietary changes or ketone supplementation, may reduce airway hyperresponsiveness and inflammation. The primary ketone body, β-hydroxybutyrate (BHB), produced during carbohydrate scarcity, acts via cell-surface receptors and transporters, potentially mitigating asthma symptoms. Weight loss and caloric restriction increase ketone levels, correlating with reduced inflammation and improved asthma outcomes. We hypothesized that β-hydroxybutyrate (BHB) reduces bronchoconstriction and inflammation in asthma by targeting bronchial smooth muscle. Using human bronchial smooth muscle cells (HBSMC) in vitro, we demonstrate herein that BHB suppresses IL-1β-induced pro-inflammatory cytokine production through Free Fatty Acid Receptor 3 (FFAR3) activation. These findings suggest that bronchial smooth muscle is a key target of therapeutic ketosis, supporting BHB's potential benefits in preclinical asthma models.
Collapse
Affiliation(s)
- V. Amanda Fastiggi
- Department of Medicine, Division of Pulmonary Disease and Critical Care, University of Vermont, and The Vermont Lung Center, Burlington, VT, 05405, USA
- Cellular, Molecular, and Biomedical Sciences Doctoral Program, University of Vermont, Burlington, VT, 05405, USA
| | - Madeleine M. Mank
- Department of Medicine, Division of Pulmonary Disease and Critical Care, University of Vermont, and The Vermont Lung Center, Burlington, VT, 05405, USA
| | - Matthew E. Poynter
- Department of Medicine, Division of Pulmonary Disease and Critical Care, University of Vermont, and The Vermont Lung Center, Burlington, VT, 05405, USA
| |
Collapse
|
32
|
Muhs T, Ljubojevic-Holzer S, Sattler S. Anti-inflammatory Therapies for Ischemic Heart Disease. Curr Cardiol Rep 2025; 27:57. [PMID: 39969632 PMCID: PMC11839821 DOI: 10.1007/s11886-025-02211-0] [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] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE OF REVIEW The inclusion of immunomodulatory strategies as supportive therapies in ischemic heart disease (IHD) has garnered significant support over recent years. Several such approaches appear to be unified through their ultimate target, the NLRP3 inflammasome. This review presents a brief update on immunomodulatory strategies in the continuum of conditions constituting ischemic heart disease and emphasising on the seemingly unifying mechanism of NLRP3 activation as well as modulation across these conditions. RECENT FINDINGS The NLRP3 inflammasome is a multiprotein complex assembled upon inflammatory stimulation, causing the release of pro-inflammatory cytokines and initiating pyroptosis. The NLRP3 pathway is relevant in inflammatory signalling of cardiac immune cells as well as non-immune cells in the myocardium, including cardiomyocytes, fibroblasts and endothelial cells. In addition to a focus on clinical outcome and efficacy trials of targeting NLRP3-related pathways, the potential connection between immunomodulation in cardiology and the NLRP3 pathway is currently being explored in preclinical trials. Colchicine, cytokine-based approaches and SGLT2 inhibitors have emerged as promising agents. However, the conditions comprising IHD including atherosclerosis, coronary artery disease (CAD), myocardial infarction (MI) and ischemic cardiomyopathy/heart failure (iCMP/HF) are not equally amenable to immunomodulation with the respective drugs. Atherosclerosis, coronary artery disease and ischemic cardiomyopathy are affected by chronic inflammation, but the immunomodulatory approach to acute inflammation in the post-MI setting remains a pharmacological challenge, as detrimental and regenerative effects of myocardial inflammation are initiated in unison. The NLRP3 inflammasome lies at the center of cell mediated inflammation in IHD. Recent trial evidence has highlighted anti-inflammatory effects of colchicine, interleukin-based therapy as well as SGLT2i in IHD and that the respective drugs modulate the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Tillmann Muhs
- Department of Pharmacology, Otto-Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Senka Ljubojevic-Holzer
- Department of Cardiology, LKH Univ. Klinikum Graz, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Susanne Sattler
- Department of Pharmacology, Otto-Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria.
- Department of Cardiology, LKH Univ. Klinikum Graz, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
33
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [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: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
34
|
Chee YJ, Dalan R, Cheung C. The Interplay Between Immunity, Inflammation and Endothelial Dysfunction. Int J Mol Sci 2025; 26:1708. [PMID: 40004172 PMCID: PMC11855323 DOI: 10.3390/ijms26041708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
The endothelium is pivotal in multiple physiological processes, such as maintaining vascular homeostasis, metabolism, platelet function, and oxidative stress. Emerging evidence in the past decade highlighted the immunomodulatory function of endothelium, serving as a link between innate, adaptive immunity and inflammation. This review examines the regulation of the immune-inflammatory axis by the endothelium, discusses physiological immune functions, and explores pathophysiological processes leading to endothelial dysfunction in various metabolic disturbances, including hyperglycemia, obesity, hypertension, and dyslipidaemia. The final section focuses on the novel, repurposed, and emerging therapeutic targets that address the immune-inflammatory axis in endothelial dysfunction.
Collapse
Affiliation(s)
- Ying Jie Chee
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138632, Singapore
| |
Collapse
|
35
|
Morshedbak M, Rahimi K, Tabandeh MR. Effect of fecal microbiota transplantation on ulcerative colitis model in rats: The gut-brain axis. Heliyon 2025; 11:e42430. [PMID: 39995913 PMCID: PMC11848074 DOI: 10.1016/j.heliyon.2025.e42430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/26/2025] Open
Abstract
Study objectives The impact of fecal microbiota transplantation (FMT) on the TLR4/MYD88/NF-kB signaling pathway in the colon in the ulcerative colitis model, as well as the incidence of anxiety behaviors caused by the colitis model was investigated. Methods Twenthy four ats were induced with ulcerative colitis using a 4 % acetic acid solution administered intrarectally and were subsequently treated with prednisolone and FMT. The study examined several indicators, such as TLR4, MYD88, and NF-κB mRNA expression, along with oxidative stress factors. Additionally, it examined the relationship between anxiety-related behaviors and colitis and assessed the pro-inflammatory cytokines in the hippocampus. Results FMT led to lower disease score index and improved colon tissue pathology findings. This was associated with reduced mRNA expression of TLR4, MYD88, and NF-κB, as well as lower levels of TOS, and higher levels of TAC, GSH, and GSSG in colon tissues. FMT was found to reduce anxiety in both the open field and elevated plus maze tests. Additionally, levels of IL-6 and TNF-a were decreased in the hippocampus. Conclusions FMT suppressed acetic acid-induced colitis by inhibiting the TLR4/MYD88/NF-kB signaling pathway. FMT reduced anxiety in open field and plus maze tests, and resulted in decreased levels of IL-6 and TNF-a in the hippocampus.
Collapse
Affiliation(s)
- Mahdis Morshedbak
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Kaveh Rahimi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohammad Reza Tabandeh
- Department of Basic Sciences, Division of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran Univeristy of Ahvaz, Ahvaz, Iran
- Stem Cells and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
36
|
Wagner CA. Beyond SGLT2: proximal tubule transporters as potential drug targets for chronic kidney disease. Nephrol Dial Transplant 2025; 40:i18-i28. [PMID: 39907544 DOI: 10.1093/ndt/gfae211] [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: 05/02/2024] [Indexed: 02/06/2025] Open
Abstract
The kidneys produce daily about 180 liters of urine but only about 2 liters are excreted. The proximal tubule plays an important role in reabsorbing the majority of filtered urine and many metabolites such as sugars, amino acids, salts or phosphate that are contained in this large volume. Reabsorption of these important metabolites is mediated by a diverse group of highly specialized transport proteins. Another group of transport proteins in the proximal tubule is responsible for the active secretion of metabolic waste products or toxins and drugs into urine. All these transporters have in common that they are directly linked to kidney metabolism and indirectly to whole-body metabolism and functions. In recent years, it has become evident that modulation of these transporters may influence the onset, progression and consequences of kidney disease. This review summarizes recent developments in this field and discusses some examples of drugs already in clinical use or in development. The examples include inhibitors of sugar transporters (SGLT2 inhibitors) that are successfully used in patients with kidney disease, diabetes or heart failure. Likewise, indirect inhibitors (acetazolamide) of an transporter absorbing sodium in exchange for protons (NHE3) are used mostly in patients with heart failure or for prevention of high altitude disease, while direct inhibitors show promise in preclinical studies to reduce damage in episodes of acute kidney disease or high blood pressure. Modulators of transporters mediating the excretion of urate have been used in patients with gout and are also discussed to prevent kidney disease. Novel drugs in development target transporters for phosphate, amino acids, or toxin and drug excretion and may be helpful for specific conditions associated with kidney disease. The advantages and challenges associated with these (novel) drugs targeting proximal tubule transport are discussed. ABSTRACT The proximal tubule is responsible for reabsorbing about 60% of filtered solutes and water and is critical for the secretion of metabolic waste products, drugs and toxins. A large number of highly specialized ion channels and transport proteins belonging to the SLC and ABC transporter families are involved. Their activity is directly or indirectly linked to ATP consumption and requires large quantities of energy and oxygen supply. Moreover, the activity of these transporters is often coupled to the movement of Na+ ions thus influencing also salt and water balance, as well as transport and regulatory processes in downstream segments. Because of their relevance for systemic ion balance, for renal metabolism or for affecting regulatory processes, proximal tubule transporters are attractive targets for existing drug and for novel strategies to reduce kidney disease progression or to alleviate the consequences of decreased kidney function. In this review, the relevance of some major proximal tubule transport systems as drug targets in individuals with chronic kidney disease (CKD) is discussed. Inhibitors of the sodium-glucose cotransporter 2, SGLT2, are now part of standard therapy in patients with CKD and/or heart failure. Also, indirect inhibition of Na+/H+-exchangers by carbonic anhydrase inhibitors and uricosuric drugs have been used for decades. Inhibition of phosphate and amino acid transporters have recently been proposed as novel principles to remove excess phosphate or to protect the proximal tubule metabolically, respectively. In addition, organic cation and anion transporters involved in drug and toxin excretion may serve as targets of new drugs. The advantages and challenges associated with (novel) drugs targeting proximal tubule transport are discussed.
Collapse
Affiliation(s)
- Carsten A Wagner
- University of Zurich - Institute of Physiology, Zurich, Switzerland
| |
Collapse
|
37
|
Zhou C, Qu S. Application and Mechanism of Action of a Ketogenic Diet in Antiepileptic Therapy. ACS Chem Neurosci 2025; 16:284-291. [PMID: 39787038 DOI: 10.1021/acschemneuro.4c00695] [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] [Indexed: 01/12/2025] Open
Abstract
Epilepsy is a chronic neurological disorder caused by abnormal discharges of neurons in the brain, which seriously affects the quality of life of patients. Although there are various drug treatments available, many epilepsy patients still experience seizures with the effect of drugs and develop refractory epilepsy. The ketogenic diet can treat drug-refractory epilepsy by regulating the body's metabolism and can enhance the quality of life by improving their cognition, behavior, and sleep quality. However, there is no unified conclusion on the mechanism through which the ketogenic diet plays a therapeutic role in epilepsy. This article provides a review of the possible mechanisms of how the ketogenic diet exerts a protective effect on epilepsy.
Collapse
Affiliation(s)
- Chang Zhou
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong 510515, P.R. China
| | - Shaogang Qu
- Department of Neurology, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
38
|
Cheon J, Kwon S, Kim M. Exerkines mitigating Alzheimer's disease progression by regulating inflammation: Focusing on macrophage/microglial NLRP3 inflammasome pathway. Alzheimers Dement 2025; 21:e14432. [PMID: 39641407 PMCID: PMC11848186 DOI: 10.1002/alz.14432] [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: 08/23/2024] [Revised: 10/16/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Recent research highlights the critical role of inflammation in accelerating amyloid beta and phosphorylated tubulin-associated protein tau cascade and Alzheimer's disease (AD) progression. Emerging evidence suggests that exercise influences AD by modulating inflammatory responses. We conducted a comprehensive search across multiple online databases. Our approach focused on previous and recent studies exploring the links among inflammation, AD, and the effects of exercise, specifically targeting research articles and books published in English. We pointed out that inflammation extends from the periphery to the central nervous system, facilitated by macrophage/microglial NLRP3 (nucleotide-binding domain, leucine rich-containing family, pyrin domain-containing protein 3) inflammasome signaling, which exacerbates classical AD mechanisms. Moreover, we provided further insights into the modulation of inflammasome signaling through exercise and exerkines, which may contribute to mitigating AD development. These insights deepen our understanding of AD mechanisms and offer the potential for identifying key therapeutic targets and biomarkers crucial for effective disease management and treatment. HIGHLIGHTS: Inflammation is potentially linked to the acceleration of classical Alzheimer's disease (AD) pathogenesis, including the pathways involving amyloid beta and phosphorylated tau, mediated by pro-inflammatory cytokines. Inflammation, initiated by the nucleotide-binding domain, leucine rich-containing family, pyrin domain-containing protein 3 (NLRP3) inflammasome signaling pathway within M1-type macrophages/microglia, may contribute to neuroinflammation and AD progression. Exercise has the potential to reduce inflammation and the development of AD by influencing NLRP3 inflammasome signaling via exerkines.
Collapse
Affiliation(s)
- Jaehwan Cheon
- Department of Biomedical ScienceKorea University College of MedicineSeongbuk‐guSeoulRepublic of Korea
- Uimyung Research Institute for NeuroscienceDepartment of PharmacySahmyook UniversityNowon‐guSeoulRepublic of Korea
| | - Soonyong Kwon
- Uimyung Research Institute for NeuroscienceDepartment of PharmacySahmyook UniversityNowon‐guSeoulRepublic of Korea
- Department of Chemistry & Life ScienceSahmyook UniversityNowon‐guSeoulRepublic of Korea
| | - Mikyung Kim
- Uimyung Research Institute for NeuroscienceDepartment of PharmacySahmyook UniversityNowon‐guSeoulRepublic of Korea
- Department of Chemistry & Life ScienceSahmyook UniversityNowon‐guSeoulRepublic of Korea
| |
Collapse
|
39
|
Kuriakose BB, Zwamel AH, Mutar AA, Uthirapathy S, Bishoyi AK, Naidu KS, Hjazi A, Nakash P, Arya R, Almalki SG. The critical role of NLRP3 in drug resistance of cancers: Focus on the molecular mechanisms and possible therapeutics. Semin Oncol 2025; 52:27-40. [PMID: 40037148 DOI: 10.1016/j.seminoncol.2025.152337] [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: 12/24/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 03/06/2025]
Abstract
Nod-like receptor protein 3 (NLRP3) is a member of the leucine-rich repeat-containing protein (NLR) canonical inflammasome family. It regulates the pathophysiology of cancer by facilitating immune responses and apoptotic proteins. Furthermore, it has been observed that chemotherapy activates NLRP3 in human malignancies. The secretion of IL-1β and IL-22 to promote cancer spread may be triggered by NLRP3 activation. Furthermore, earlier studies have exhibited that NLRP3 may cause medication resistance when used in cancer treatments given that cell viability may be regulated by NLRP3 depletion. Additionally, clinical studies have demonstrated correlation between NLRP3 expression, lymphogenesis, and cancer metastasis. Various NLRP3 agonists may cause the EMT process, stimulate IL-1β and Wnt/β-catenin signaling, and alter miRNA function in drug-resistant cells. This review seeks to clarify the possibility involvement of NLRP3-related pathways in the control of cancer cells' resistance to widely used treatment approaches, such as chemotherapy. In the end, an improved perception of the corresponding mechanisms behind NLRP3's tumor-supporting activities will help NLRP3-based treatments advance in the future.
Collapse
Affiliation(s)
- Beena Briget Kuriakose
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King khalid University, Khamis Mushayt, Kingdom of Saudi Arabia
| | - Ahmed Hussein Zwamel
- Department of medical analysis, Medical laboratory technique college, the Islamic University, Najaf, Iraq; Department of medical analysis, Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Department of medical analysis, Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| | - Ayad Abdulrazzaq Mutar
- Medical Laboratory Techniques department, College of Health and medical technology, Al-maarif University, Anbar, Iraq.
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Princse Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Prashant Nakash
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, India
| | - Renu Arya
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Saudi Arabia
| |
Collapse
|
40
|
Albers GJ, Michalaki C, Ogger PP, Lloyd AF, Causton B, Walker SA, Caldwell A, Halket JM, Sinclair LV, Forde SH, McCarthy C, Hinks TSC, Lloyd CM, Byrne AJ. Airway macrophage glycolysis controls lung homeostasis and responses to aeroallergen. Mucosal Immunol 2025; 18:121-134. [PMID: 39426627 DOI: 10.1016/j.mucimm.2024.10.002] [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: 04/17/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/21/2024]
Abstract
The lungs represent a dynamic microenvironment where airway macrophages (AMs) are the major lung-resident macrophages. AMs dictate the balance between tissue homeostasis and immune activation and thus have contradictory functions by maintaining tolerance and tissue homeostasis, as well as initiating strong inflammatory responses. Emerging evidence has highlighted the connection between macrophage function and cellular metabolism. However, the functional importance of these processes in tissue-resident specialized macrophage populations such as those found in the airways, remain poorly elucidated. Here, we reveal that glycolysis is a fundamental pathway in AMs which regulates both lung homeostasis and responses to inhaled allergen. Using macrophage specific targeting in vivo, and multi-omics approaches, we determined that glycolytic activity in AMs is necessary to restrain type 2 (T2) immunity during homeostasis. Exposure to a range of common aeroallergens, including house dust mite (HDM), drove AM-glycolysis and furthermore, AM-specific inhibition of glycolysis altered inflammation in the airways and HDM-driven airway metabolic adaptations in vivo. Additionally, allergen sensitised asthmatics had profound metabolic changes in the airways, compared to non-sensitised asthmatic controls. Finally, we found that allergen driven AM-glycolysis in mice was TLR2 dependent. Thus, our findings demonstrate a direct relationship between glycolysis in AMs, AM-mediated homeostatic processes, and T2 immune responses in the lungs. These data suggest that glycolysis is essential for the plasticity of AMs. Depending on the immunological context, AM-glycolysis is required to exert homeostatic activity but once activated by allergen, AM-glycolysis influences inflammatory responses. Thus, precise modulation of glycolytic activity in AMs is essential for preserving lung homeostasis and regulating airway inflammation.
Collapse
Affiliation(s)
- Gesa J Albers
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Patricia P Ogger
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Amy F Lloyd
- Cell Signalling and Immunology, University of Dundee, Dundee, UK
| | - Benjamin Causton
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Simone A Walker
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Anna Caldwell
- Dept. of Nutritional Sciences, School of Life Course & Population Health Sciences, King's College London, London, UK; Department of Nutritional Sciences, KIng's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - John M Halket
- Department of Nutritional Sciences, KIng's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Linda V Sinclair
- Cell Signalling and Immunology, University of Dundee, Dundee, UK
| | - Sarah H Forde
- Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | - Cormac McCarthy
- Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | - Timothy S C Hinks
- Respiratory Medicine Unit, Nuffield Department of Medicine and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, and the NIHR Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, UK
| | - Clare M Lloyd
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Adam J Byrne
- National Heart and Lung Institute, Imperial College London, London, UK; Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland.
| |
Collapse
|
41
|
Tork MAB, Fotouhi S, Roozi P, Negah SS. Targeting NLRP3 Inflammasomes: A Trojan Horse Strategy for Intervention in Neurological Disorders. Mol Neurobiol 2025; 62:1840-1881. [PMID: 39042218 DOI: 10.1007/s12035-024-04359-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Recently, a growing focus has been on identifying critical mechanisms in neurological diseases that trigger a cascade of events, making it easier to target them effectively. One such mechanism is the inflammasome, an essential component of the immune response system that plays a crucial role in disease progression. The NLRP3 (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 3) inflammasome is a subcellular multiprotein complex that is widely expressed in the central nervous system (CNS) and can be activated by a variety of external and internal stimuli. When activated, the NLRP3 inflammasome triggers the production of proinflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18) and facilitates rapid cell death by assembling the inflammasome. These cytokines initiate inflammatory responses through various downstream signaling pathways, leading to damage to neurons. Therefore, the NLRP3 inflammasome is considered a significant contributor to the development of neuroinflammation. To counter the damage caused by NLRP3 inflammasome activation, researchers have investigated various interventions such as small molecules, antibodies, and cellular and gene therapy to regulate inflammasome activity. For instance, recent studies indicate that substances like micro-RNAs (e.g., miR-29c and mR-190) and drugs such as melatonin can reduce neuronal damage and suppress neuroinflammation through NLRP3. Furthermore, the transplantation of bone marrow mesenchymal stem cells resulted in a significant reduction in the levels of pyroptosis-related proteins NLRP3, caspase-1, IL-1β, and IL-18. However, it would benefit future research to have an in-depth review of the pharmacological and biological interventions targeting inflammasome activity. Therefore, our review of current evidence demonstrates that targeting NLRP3 inflammasomes could be a pivotal approach for intervention in neurological disorders.
Collapse
Affiliation(s)
- Mohammad Amin Bayat Tork
- Clinical Research Development Unit, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soroush Fotouhi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Roozi
- Department of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sajad Sahab Negah
- Clinical Research Development Unit, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran.
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Pardis Campus, Azadi Square, Kalantari Blvd., Mashhad, Iran.
| |
Collapse
|
42
|
Ong LT, Sia CH. Interactions between antidiabetes medications and heart-brain axis. Curr Opin Endocrinol Diabetes Obes 2025; 32:34-43. [PMID: 39639832 DOI: 10.1097/med.0000000000000896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
PURPOSE OF REVIEW The heart - brain axis (HBA) is the physiological interactions between the cardiovascular and nervous systems through autonomic nerves, hormones, and cytokines. Patients diagnosed with diabetes mellitus have an increased risk of the cardiovascular and neurological diseases. However, recent evidence demonstrated that different antidiabetic drugs may delay cognitive impairment and improve cardiovascular outcomes. This review examines the impact of antidiabetic drugs on the HBA in patients with diabetes. RECENT FINDINGS Metformin improves the cardiovascular and cognitive outcomes through adenosine 5'-monophosphate-activated protein kinase activation. Sodium-glucose cotransporter-2 inhibitors reduce inflammation, oxidative stress by inhibiting the NLRP3 inflammasome thereby reducing the incidence of heart failure and formation of beta-amyloid and neurofibrillary tangles in the brain. Dipeptidyl peptidase-4 inhibitors exhibit neuroprotective effects in Alzheimer's disease by reducing amyloid-beta and tau pathology and inflammation but may exacerbate heart failure risk due to increased sympathetic activity and prolonged β-adrenergic stimulation. Glucagon-like peptide-1 receptor agonists exhibit neuroprotective effects in Alzheimer's and Parkinson's diseases by reducing neuroinflammation, but may increase sympathetic activity, potentially elevating heart rate and blood pressure, despite their cardioprotective benefits. SUMMARY Antidiabetes medications have the potential to improve cardiovascular and cognitive outcomes; however, additional studies are required to substantiate these effects.
Collapse
Affiliation(s)
- Leong Tung Ong
- Department of Cardiology, National University Heart Centre, Singapore
| | | |
Collapse
|
43
|
Miura A, Yamanashi T, Kajitani N, Fukuda S, Tsunetomi K, Matsuo R, Nishiguchi T, Pu S, Nakada Y, Shirayama Y, Watanabe K, Kaneko K, Iwata M. Medium-Chain Triglyceride Administration Induces Antidepressant Effects in Animal Models by Increasing Beta-Hydroxybutyrate Levels. Yonago Acta Med 2025; 68:58-67. [PMID: 39968118 PMCID: PMC11831044 DOI: 10.33160/yam.2025.02.007] [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: 11/29/2024] [Accepted: 12/28/2024] [Indexed: 02/20/2025]
Abstract
Background Inflammation is believed to contribute to the pathophysiology of depression, with increased levels of inflammatory cytokines, such as interleukin-1β (IL-1β), observed in patients. Depression is also common in individuals with chronic inflammatory diseases. IL-1β disrupts synaptic transmission and reduces neurogenesis in the hippocampus, playing a crucial role in depression development. Our prior research found that stress activates microglia in the brain to produce IL-1β via the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome. Additionally, β-hydroxybutyrate (BHB), an endogenous ketone body, alleviates stress-induced depression by inhibiting NLRP3 activation and IL-1β production. However, BHB's poor bioavailability limits its effectiveness. Medium-chain triglycerides (MCTs) can increase blood BHB levels, making them a potential treatment for stress-induced depression. Methods We tested MCT in two animal models: social defeat (SD) in mice and chronic unpredictable stress (CUS) in rats. MCT was orally administered to both groups to assess blood BHB levels. Behavioral tests, including the forced swim test (FST), were performed, and brain tissue was analyzed for IL-1β levels and spine density. Results MCT administration increased blood BHB levels 7-11 times within 1 hour. In the SD model, MCT significantly reduced immobility time in the FST, suggesting antidepressant effects. While the CUS model showed no significant change, a trend toward reduced immobility time was observed. MCT treatment also reduced stress-induced IL-1β levels in the rat hippocampus, although spine density remained unchanged. Conclusion MCT appears to alleviate stress-induced depression-like behaviors, likely through the suppression of IL-1β in the hippocampus. Owing to its ease of oral administration, MCT may offer a practical treatment for stress-related depression.
Collapse
Affiliation(s)
- Akihiko Miura
- Division of Neuropsychiatry, Department of Brain and Neurosciences, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Takehiko Yamanashi
- Division of Neuropsychiatry, Department of Brain and Neurosciences, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Naofumi Kajitani
- Division of Neuropsychiatry, Department of Brain and Neurosciences, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Saki Fukuda
- Division of Neuropsychiatry, Department of Brain and Neurosciences, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Kyohei Tsunetomi
- Division of Neuropsychiatry, Department of Brain and Neurosciences, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Ryoichi Matsuo
- Division of Neuropsychiatry, Department of Brain and Neurosciences, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Tsuyoshi Nishiguchi
- Division of Neuropsychiatry, Department of Brain and Neurosciences, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Shenghong Pu
- Division of Neuropsychiatry, Department of Brain and Neurosciences, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Yumeto Nakada
- Division of Clinical Laboratory, Tottori University Hospital, Yonago 683-8504, Japan
| | - Yukihiko Shirayama
- Department of Psychiatry, Teikyo University Chiba Medical Center, Ichihara 299-0112, Japan
| | | | - Koichi Kaneko
- Division of Neuropsychiatry, Department of Brain and Neurosciences, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Masaaki Iwata
- Division of Neuropsychiatry, Department of Brain and Neurosciences, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| |
Collapse
|
44
|
Huizer K, Soni S, Schmidt MA, Çakici N, de Haan L, Dyck JRB, van Beveren NJM. Potential benefits of ketone therapy as a novel immunometabolic treatment for schizophrenia. Psychiatry Res 2025; 345:116379. [PMID: 39892306 DOI: 10.1016/j.psychres.2025.116379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 11/14/2024] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Therapeutic ketosis could target the potential bio-energetic pathophysiology of schizophrenia. Ideally, novel treatments also target the possible inflammatory aspects of schizophrenia. Adult mice (n = 30) were treated with ketone ester (KE) or vehicle for 3 days, next LPS- or PBS-injected. Brains were collected the next day. KE significantly attenuated the increased transcription of the pro-inflammatory cytokines Tnf-a, Il-6 and Il-1b, without affecting anti-inflammatory/immunomodulatory cytokines (Il-4, Il-10, Il-11) in whole brain. KE potently dampened neuro-inflammation in this acute inflammation mouse model. Ketone therapy could simultaneously target two possible pathophysiological pathways in schizophrenia. We encourage more research into the immunometabolic potential of therapeutic ketosis in schizophrenia.
Collapse
Affiliation(s)
- Karin Huizer
- Parnassia Academy, Parnassia Psychiatric Institute, The Hague, the Netherlands; Department of Psychiatry and Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands; Department of Pathology, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - Shubham Soni
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Mya A Schmidt
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Nuray Çakici
- Parnassia Academy, Parnassia Psychiatric Institute, The Hague, the Netherlands; Department of Psychiatry and Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Lieuwe de Haan
- Department of Psychiatry and Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Jason R B Dyck
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
45
|
Lv T, Liu C, Guo S, Wu M, Wang X, Zhang Z, Zhou J, Yao Y, Shen Z, Yang J, Sun S, Liu Z, Chi J. Targeting Ketone Body Metabolism Improves Cardiac Function and Hemodynamics in Patients With Heart Failure: A Systematic Review and Meta-Analysis. Nutr Rev 2025:nuae179. [PMID: 39873669 DOI: 10.1093/nutrit/nuae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
CONTEXT The impacts of elevated ketone body levels on cardiac function and hemodynamics in patients with heart failure (HF) remain unclear. OBJECTIVE The effects of ketone intervention on these parameters in patients with HF were evaluated quantitatively in this meta-analysis. DATA SOURCES We searched the PubMed, Cochrane Library, and Embase databases for relevant studies published from inception to April 13, 2024. Ketone therapy included ketone ester and β-hydroxybutyrate intervention. DATA EXTRACTION Seven human studies were included for the quantitative analysis. DATA ANALYSIS Our results showed that ketone therapy significantly improved left ventricular ejection fraction (standardized mean difference, 0.52 [95% CI, 0.25-0.80]; I2 = 0%), cardiac output (0.84 [95% CI, 0.36-1.32]; I2 = 68%) and stroke volume (0.47 [95% CI, 0.10-0.84]; I2 = 39%), and significantly reduced systemic vascular resistance (-0.92 [95% CI, -1.52 to -0.33]; I2 = 74%) without influencing mean arterial pressure (-0.09 [95% CI: -0.40 to 0.22]; I2 = 0%) in patients with HF. Subgroup analysis revealed that the enhanced cardiac function and favorable hemodynamic effects of ketone therapy were also applicable to individuals without HF. CONCLUSIONS Ketone therapy may significantly improve cardiac systolic function and hemodynamics in patients with HF and in patients without HF, suggesting it may be a promising treatment for patients with HF and also a beneficial medical strategy for patients without HF or healthy individuals.
Collapse
Affiliation(s)
- Tingting Lv
- Department of General Practice, Shaoxing People's Hospital, Shaoxing 312000, P. R. China
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Chunyan Liu
- Department of Infection Management, Shaoxing People's Hospital, Shaoxing 312000, P. R. China
| | - Shitian Guo
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P. R. China
| | - Menglu Wu
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Xiang Wang
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Ziyi Zhang
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Jiedong Zhou
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Yiying Yao
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Zeyu Shen
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Juntao Yang
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Shijia Sun
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Zheng Liu
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Jufang Chi
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Department of Cardiology, Zhuji People's Hospital (Zhuji Hospital, Wenzhou Medical University), Zhuji, Zhejiang 311800, P. R. China
| |
Collapse
|
46
|
Avram L, Crișan D, Moldovan RC, Bogos LG, Iuga CA, Andraș D, Crișan S, Bodolea C, Nemeş A, Donca V. Metabolomic Exploration of Colorectal Cancer Through Amino Acids and Acylcarnitines Profiling of Serum Samples. Cancers (Basel) 2025; 17:427. [PMID: 39941796 PMCID: PMC11816151 DOI: 10.3390/cancers17030427] [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: 12/17/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES Colorectal cancer (CRC) represents one of the most prevalent forms of cancer, with high mortality rates. The aim of this study was to observe and understand the metabolic changes in CRC through targeted metabolomics. METHODS Samples collected from 58 CRC patients and 35 healthy individuals have been analyzed by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS), targeting two classes of metabolites: amino acids and acylcarnitines. RESULTS Statistical analysis revealed 26 significantly modified (p-value < 0.01; |FC| > 1.2) metabolites in CRC patients compared to the control group and 22 between colon cancer and control, whereas 8 metabolites differed only significantly between rectal cancer and healthy patients. Some of these significantly modified metabolites characterize cancer-specific adaptations, such as increased energy demand, increased tumor invasiveness, capabilities to promote amino acid synthesis, and tumor resistance against acute immune response. Moreover, receiver operator characteristic (ROC) analysis revealed that a set of two acylcarnitines (C6DC and C4-OH) can differentiate between CRC patients and healthy individuals with a high degree of confidence (AUC 0.837). CONCLUSIONS By implementing a metabolomics approach targeting amino acids and acylcarnitines, several metabolic alterations induced by CRC have been highlighted. Even though these modifications are not specific enough to act as disease markers, they might prove useful for evaluating patient status.
Collapse
Affiliation(s)
- Lucreția Avram
- Geriatrics—Gerontology, Department 5—Medical Specialties, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (L.A.); (V.D.)
| | - Dana Crișan
- Department of Internal Medicine, 5th Medical Clinic, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (D.C.); (S.C.)
| | - Radu-Cristian Moldovan
- Department of Personalized Medicine and Rare Diseases, Institute of Biomedical Research—MedFuture, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (L.-G.B.); (C.-A.I.)
| | - Luisa-Gabriela Bogos
- Department of Personalized Medicine and Rare Diseases, Institute of Biomedical Research—MedFuture, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (L.-G.B.); (C.-A.I.)
| | - Cristina-Adela Iuga
- Department of Personalized Medicine and Rare Diseases, Institute of Biomedical Research—MedFuture, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (L.-G.B.); (C.-A.I.)
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - David Andraș
- 1st Surgical Clinic, Department of General Surgery, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj Napoca, Romania;
| | - Sorin Crișan
- Department of Internal Medicine, 5th Medical Clinic, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (D.C.); (S.C.)
| | - Constantin Bodolea
- Intensive Care Unit Department, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (C.B.); (A.N.)
| | - Andrada Nemeş
- Intensive Care Unit Department, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (C.B.); (A.N.)
| | - Valer Donca
- Geriatrics—Gerontology, Department 5—Medical Specialties, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy Cluj-Napoca, 400012 Cluj-Napoca, Romania; (L.A.); (V.D.)
| |
Collapse
|
47
|
Li D, Zhang L, Gong Q, Deng H, Luo C, Zhou T, Huang W, Xu Y. The role of myocardial energy metabolism perturbations in diabetic cardiomyopathy: from the perspective of novel protein post-translational modifications. Clin Epigenetics 2025; 17:15. [PMID: 39865334 PMCID: PMC11765930 DOI: 10.1186/s13148-025-01814-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/06/2025] [Indexed: 01/28/2025] Open
Abstract
Diabetic cardiomyopathy (DbCM), a significant chronic complication of diabetes, manifests as myocardial hypertrophy, fibrosis, and other pathological alterations that substantially impact cardiac function and elevate the risk of cardiovascular diseases and patient mortality. Myocardial energy metabolism disturbances in DbCM, encompassing glucose, fatty acid, ketone body and lactate metabolism, are crucial factors that contribute to the progression of DbCM. In recent years, novel protein post-translational modifications (PTMs) such as lactylation, β-hydroxybutyrylation, and succinylation have been demonstrated to be intimately associated with the myocardial energy metabolism process, and in conjunction with acetylation, they participate in the regulation of protein activity and gene expression activity in cardiomyocytes. This review examines the epigenetic pathogenesis of DbCM, primarily focusing on myocardial energy metabolism perturbations and novel PTMs associated with them. It provides a detailed analysis of the mechanisms of these novel PTMs in DbCM to enhance the understanding of DbCM pathophysiology and establish a theoretical foundation for the development of new treatment strategies for DbCM.
Collapse
Affiliation(s)
- Dongze Li
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, 646000, Sichuan, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China
| | - Li Zhang
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, 646000, Sichuan, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China
- Department of Du's Orthopedic Surgery, Sichuan Second Hospital of Traditional Chinese Medicine, Chengdu, 610000, Sichuan, China
| | - Qiming Gong
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Nephrology, Youjiang Medical College for Nationalities Affiliated Hospital, Youjiang, 533000, Guangxi, China
- Guangxi Key Laboratory of Basic Medical Research Support for Immune Related Diseases, Youjiang Medical University for Nationalities, Youjiang, 533000, Guangxi, China
| | - Huilan Deng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, 646000, Sichuan, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China
| | - Changfang Luo
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, 646000, Sichuan, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China
| | - Tingting Zhou
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, 646000, Sichuan, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China
| | - Wei Huang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, 646000, Sichuan, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China.
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Sichuan Clinical Research Center for Diabetes and Metabolic Diseases, Luzhou, 646000, Sichuan, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
48
|
Ji J, Tang Y. A ketogenic diet regulates microglial activation to treat drug addiction. Front Pharmacol 2025; 16:1462699. [PMID: 39917617 PMCID: PMC11799558 DOI: 10.3389/fphar.2025.1462699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025] Open
Abstract
Drug addiction is a chronic and potentially deadly disease that is considered a global health problem and describes the alteration of brain function by psychostimulant drugs through changes in the reward system. However, there is still no ideal strategy for the management of drug addiction. Previous studies have suggested that microglia are involved in events associated with neuroplasticity and memory, which are also related to drug addiction. Many studies have shown that psychoactive substances may act directly on immune cells, altering their function and inducing the production of various inflammatory mediators. In recent years, a ketogenic diet (KD) was shown to have therapeutic benefits as a dietary therapy for a variety of neurological disorders. With respect to drug addiction, studies have shown that a KD can alleviate glucose metabolism disorders caused by alcohol use disorders by increasing ketone metabolism, thereby reducing withdrawal symptoms. This finding indicates the potential of a KD as a treatment for drug addiction, since a KD may promote the transition of microglia to a predominantly anti-inflammatory state through several mechanisms. Here, we discuss recent research showing that a KD plays a variety of roles in controlling microglia-mediated inflammation, opening new treatment avenues to treat drug addiction. This succinct analysis offers evidence of the enormous potential of a KD to treat drug addiction through the inhibition of microglial activation.
Collapse
Affiliation(s)
- Jie Ji
- Department of Comprehensive (VIP) Inpatient Ward, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Tang
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
49
|
Mao J, Xia W, Wu Y, Li M, Zhao Y, Zhai P, Zhang Y, Zan T, Cui W, Sun X. Biosynthesis of Lysosomally Escaped Apoptotic Bodies Inhibits Inflammasome Synthesis in Macrophages. RESEARCH (WASHINGTON, D.C.) 2025; 8:0581. [PMID: 39850366 PMCID: PMC11754539 DOI: 10.34133/research.0581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/07/2024] [Accepted: 12/22/2024] [Indexed: 01/25/2025]
Abstract
Hyperglycemia and bacterial colonization in diabetic wounds aberrantly activate Nod-like receptor protein 3 (NLRP3) in macrophages, resulting in extensive inflammatory infiltration and impaired wound healing. Targeted suppression of the NLRP3 inflammasome shows promise in reducing macrophage inflammatory disruptions. However, challenges such as drug off-target effects and degradation via lysosomal capture remain during treatment. In this study, engineered apoptotic bodies (BHB-dABs) derived from adipose stem cells loaded with β-hydroxybutyric acid (BHB) were synthesized via biosynthesis. These vesicles target M1-type macrophages, which highly express the folic acid receptor in the inflammatory microenvironment, and facilitate lysosomal escape through 1,2-distearoyl-sn-propyltriyl-3-phosphatidylethanolamine-polyethylene glycol functionalization, which may enhance the efficacy of NLRP3 inhibition for managing diabetic wounds. In vitro studies demonstrated the biocompatibility of BHB-dABs, their selective targeting of M1-type macrophages, and their ability to release BHB within the inflammatory microenvironment via folic acid and folic acid receptor signaling. These nanovesicles exhibited lysosomal escape, anti-inflammatory, mitochondrial protection, and endothelial cell vascularization properties. In vivo experiments demonstrated that BHB-dABs enhance the recovery of diabetic wound inflammation and angiogenesis, accelerating wound healing. These functionalized apoptotic bodies efficiently deliver NLRP3 inflammasome inhibitors using a dual strategy of targeting macrophages and promoting lysosomal escape. This approach represents a novel therapeutic strategy for effectively treating chronic diabetic wounds.
Collapse
Affiliation(s)
- Jiayi Mao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Wenzheng Xia
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Yanglin Wu
- Department of Orthopaedics, Shanghai Tenth People’s Hospital,
Tongji University School of Medicine, Shanghai 200072, P. R. China
| | - Minxiong Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Yun Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Peisong Zhai
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuguang Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Xiaoming Sun
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| |
Collapse
|
50
|
Liang IC, Chang HH, Lai YJ, Chan CM, Sung CH, Pu CM, Chang DC, Ho CC, Hung CF. Update on the Efficacy and Safety of Sodium-Glucose Co-Transporter 2 Inhibitors in Patients with Chronic Diseases: A Systematic Review and Meta-Analysis. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:202. [PMID: 40005319 PMCID: PMC11857657 DOI: 10.3390/medicina61020202] [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: 12/25/2024] [Revised: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 02/27/2025]
Abstract
Background: Sodium-glucose co-transporter-2 (SGLT2) inhibitors have emerged as vital medications for the management of type 2 diabetes mellitus (T2DM). Numerous studies have highlighted the cardioprotective and renal protective benefits of SGLT2 inhibitors. Consequently, it is essential to assess their efficacy and safety in patients with chronic diseases. Method: We conducted a systematic review and meta-analysis of randomized controlled trials (RCTs) evaluating the effects of SGLT2 inhibitors on major cardiovascular and safety outcomes in patients with T2DM, heart failure (HF), and chronic kidney disease (CKD). We searched the PubMed, Cochrane, and Embase databases for trials published between 30 September 2021 and 17 May 2023. The primary outcomes of interest included nonfatal myocardial infarction (MI), hospitalization for heart failure (HHF), cardiovascular death, and nonfatal stroke. The safety outcomes assessed were hypoglycemia, urinary tract infections (UTIs), and acute kidney injury (AKI). Result: We identified 13 RCTs involving 90,413 participants. In patients with T2DM, SGLT2 inhibitors significantly reduced the risk of nonfatal MI by 12% (hazard ratio [HR] = 0.88, 95% confidence interval [CI]: 0.78-0.98), HHF by 33% (HR = 0.67, 95% CI: 0.62-0.74), and cardiac death by 15% (HR = 0.95, 95% CI: 0.80-1.13). However, they did not significantly reduce the risk of nonfatal stroke (HR = 0.85, 95% CI: 0.75-0.95). In patients with HF, SGLT2 inhibitors reduced the risk of HHF by 28% (HR = 0.72, 95% CI: 0.66-0.77) and cardiac death by 12% (HR = 0.88, 95% CI: 0.80-0.96). For patients with CKD, SGLT2 inhibitors reduced the risk of HHF by 35% (HR = 0.65, 95% CI: 0.55-0.76) and cardiac death by 16% (HR = 0.84, 95% CI: 0.73-0.96). Regarding safety outcomes, SGLT2 inhibitors did not significantly increase the risk of hypoglycemia in patients with T2DM, HF, or CKD, nor did they increase the risk of urinary tract infections (UTIs) in patients with HF or CKD, or the risk of acute kidney injury (AKI) in patients with HF. However, they did increase the risk of UTIs by 8% (risk ratio [RR] = 1.08, 95% CI: 1.01-1.16) in patients with T2DM and reduced the risk of AKI by 22% (RR = 0.78, 95% CI: 0.67-0.89) and 19% (RR = 0.81, 95% CI: 0.69-0.97) in patients with T2DM and CKD, respectively. Conclusions: SGLT2 inhibitors have demonstrated a significant improvement in cardiovascular outcomes for patients with T2DM, HF, and CKD while also maintaining a favorable safety profile. These findings advocate for the broader application of SGLT2 inhibitors in the management of chronic diseases, particularly in reducing the incidence of nonfatal MI, HHF, and cardiac death. Further research is essential to optimize their use across diverse patient populations and stages of disease.
Collapse
Affiliation(s)
- I-Chia Liang
- National Defense Medical Center, Department of Ophthalmology, Tri-Service General Hospital, Taipei 11490, Taiwan;
- Department of Ophthalmology, Cathay General Hospital, Taipei 10630, Taiwan
| | - Hsun-Hao Chang
- Department of Cardiology, Tainan Municipal Hospital (Managed by Show Chwan Medical Care Corporation), Tainan 70173, Taiwan;
| | - Yu-Jou Lai
- Department of Pharmacy, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City 24205, Taiwan;
| | - Chi-Ming Chan
- Department of Ophthalmology, Cardinal Tien Hospital, New Taipei City 23148, Taiwan;
| | - Chao-Hsien Sung
- Division of Anesthesiology, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City 24205, Taiwan;
| | - Chi-Ming Pu
- Division of Plastic Surgery, Department of Surgery, Cathay General Hospital, Taipei 10630, Taiwan;
| | - Der-Chen Chang
- Department of Mathematics and Statistics, Department of Computer Science, Georgetown University, Washington, DC 20057, USA;
| | - Ching-Chih Ho
- Department of Anesthesiology, Taoyuan Armed Forces General Hospital, Taoyuan 32551, Taiwan
| | - Chi-Feng Hung
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| |
Collapse
|