1
|
Flores-Hernández MN, Martínez-Coria H, López-Valdés HE, Arteaga-Silva M, Arrieta-Cruz I, Gutiérrez-Juárez R. Efficacy of a High-Protein Diet to Lower Glycemic Levels in Type 2 Diabetes Mellitus: A Systematic Review. Int J Mol Sci 2024; 25:10959. [PMID: 39456742 PMCID: PMC11507302 DOI: 10.3390/ijms252010959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Diabetes is a metabolic disease with a high worldwide prevalence and an important factor in mortality and disability in the population. Complications can be reduced or prevented with lifestyle changes in physical activity, dietary habits, and smoking cessation. High-protein diets (HPDs, >30% or >1.0 g/Kg/day) decrease hyperglycemia in part due to their content of branched-chain amino acids (BCAAs), mainly leucine. Leucine (and other BCAAs) improve glucose metabolism by directly signaling in the medio-basal hypothalamus (MBH), increasing liver insulin sensitivity. To determine the effectiveness of an HPD to lower hyperglycemia, we analyzed the results of published clinical studies focusing on the levels of fasting plasma glucose and/or glycosylated hemoglobin (HbA1c) in patients with type 2 diabetes mellitus (T2DM). We carried out a systematic search for clinical studies using HPDs. We searched five databases (Scopus, Web of Science, PubMed, Epistemonikos, and Cochrane), collecting 179 articles and finally selecting 8 articles to analyze their results. In conclusion, HPDs are an effective alternative to reduce hyperglycemia in patients with T2DM, especially so-called Paleolithic diets, due to their higher-quality protein from animal and vegetal sources and their exclusion of grains, dairy products, salt, refined fats, and added sugars.
Collapse
Affiliation(s)
- María Nelly Flores-Hernández
- Departamento de Ciencias Biomédicas, Escuela de Medicina, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Mexico City 09230, Mexico;
| | - Hilda Martínez-Coria
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico; (H.M.-C.); (H.E.L.-V.)
| | - Héctor E. López-Valdés
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico; (H.M.-C.); (H.E.L.-V.)
| | - Marcela Arteaga-Silva
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico;
| | - Isabel Arrieta-Cruz
- Departamento de Investigación Básica, División de Investigación, Instituto Nacional de Geriatría, Secretaría de Salud, Mexico City 10200, Mexico;
| | - Roger Gutiérrez-Juárez
- Departamento de Ciencias Biomédicas, Escuela de Medicina, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Mexico City 09230, Mexico;
| |
Collapse
|
2
|
Wang J, Wu Q, Wang X, Liu H, Chen M, Xu L, Zhang Z, Li K, Li W, Zhong J. Targeting Macrophage Phenotypes and Metabolism as Novel Therapeutic Approaches in Atherosclerosis and Related Cardiovascular Diseases. Curr Atheroscler Rep 2024; 26:573-588. [PMID: 39133247 PMCID: PMC11392985 DOI: 10.1007/s11883-024-01229-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 08/13/2024]
Abstract
PURPOSE OF THE REVIEW Macrophage accumulation and activation function as hallmarks of atherosclerosis and have complex and intricate dynamics throughout all components and stages of atherosclerotic plaques. In this review, we focus on the regulatory roles and underlying mechanisms of macrophage phenotypes and metabolism in atherosclerosis. We highlight the diverse range of macrophage phenotypes present in atherosclerosis and their potential roles in progression and regression of atherosclerotic plaque. Furthermore, we discuss the challenges and opportunities in developing therapeutic strategies for preventing and treating atherosclerotic cardiovascular disease. RECENT FINDINGS Dysregulation of macrophage polarization between the proinflammatory M1 and anti-inflammatory M2 phenotypealters the immuno-inflammatory response during atherosclerosis progression, leading to plaque initiation, growth, and ultimately rupture. Altered metabolism of macrophage is a key feature for their function and the subsequent progression of atherosclerotic cardiovascular disease. The immunometabolism of macrophage has been implicated to macrophage activation and metabolic rewiring of macrophages within atherosclerotic lesions, thereby shifting altered macrophage immune-effector and tissue-reparative function. Targeting macrophage phenotypes and metabolism are potential therapeutic strategies in the prevention and treatment of atherosclerosis and atherosclerotic cardiovascular diseases. Understanding the precise function and metabolism of specific macrophage subsets and their contributions to the composition and growth of atherosclerotic plaques could reveal novel strategies to delay or halt development of atherosclerotic cardiovascular diseases and their associated pathophysiological consequences. Identifying biological stimuli capable of modulating macrophage phenotypes and metabolism may lead to the development of innovative therapeutic approaches for treating patients with atherosclerosis and coronary artery diseases.
Collapse
Affiliation(s)
- Juan Wang
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Qiang Wu
- Senior Department of Cardiology, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
- Journal of Geriatric Cardiology Editorial Office, Chinese PLA General Hospital, Beijing, China
| | - Xinyu Wang
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Hongbin Liu
- Department of Cardiology, the Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Mulei Chen
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Li Xu
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ze Zhang
- National Institute of Biological Sciences, Beijing, China
| | - Kuibao Li
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Weiming Li
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Jiuchang Zhong
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Bonacina F, Zhang X, Manel N, Yvan-Charvet L, Razani B, Norata GD. Lysosomes in the immunometabolic reprogramming of immune cells in atherosclerosis. Nat Rev Cardiol 2024:10.1038/s41569-024-01072-4. [PMID: 39304748 DOI: 10.1038/s41569-024-01072-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
Lysosomes have a central role in the disposal of extracellular and intracellular cargo and also function as metabolic sensors and signalling platforms in the immunometabolic reprogramming of macrophages and other immune cells in atherosclerosis. Lysosomes can rapidly sense the presence of nutrients within immune cells, thereby switching from catabolism of extracellular material to the recycling of intracellular cargo. Such a fine-tuned degradative response supports the generation of metabolic building blocks through effectors such as mTORC1 or TFEB. By coupling nutrients to downstream signalling and metabolism, lysosomes serve as a crucial hub for cellular function in innate and adaptive immune cells. Lysosomal dysfunction is now recognized to be a hallmark of atherogenesis. Perturbations in nutrient-sensing and signalling have profound effects on the capacity of immune cells to handle cholesterol, perform phagocytosis and efferocytosis, and limit the activation of the inflammasome and other inflammatory pathways. Strategies to improve lysosomal function hold promise as novel modulators of the immunoinflammatory response associated with atherosclerosis. In this Review, we describe the crosstalk between lysosomal biology and immune cell function and polarization, with a particular focus on cellular immunometabolic reprogramming in the context of atherosclerosis.
Collapse
Affiliation(s)
- Fabrizia Bonacina
- Department of Excellence of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Xiangyu Zhang
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Nicolas Manel
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU), Oncoage, Nice, France
| | - Babak Razani
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Giuseppe D Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
4
|
Zhen LL, Feng L, Jiang WD, Wu P, Liu Y, Tang L, Li SW, Zhong CB, Zhou XQ. Exploring the novel benefits of leucine: Protecting nitrite-induced liver damage in sub-adult grass carp (Ctenopharyngodon idella) through regulating mitochondria quality control. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109690. [PMID: 38866347 DOI: 10.1016/j.fsi.2024.109690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/21/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024]
Abstract
Leucine is an essential amino acid for fish. The ability of leucine to resist stress in fish has not been reported. Nitrite is a common pollutant in the aquatic environment. Therefore, we investigated the effects of dietary leucine on growth performance and nitrite-induced liver damage, mitochondrial dysfunction, autophagy, and apoptosis for sub-adult grass carp. A total of 450 grass carp (615.91 ± 1.15 g) were selected and randomly placed into 18 net cages. The leucine contents of the six diets were 2.91, 5.90, 8.92, 11.91, 14.93, and 17.92 g/kg, respectively. After a 9-week feeding trial, the nitrite exposure experiment was set up for 96 h. These results indicated that dietary leucine significantly promoted FW, WG, PWG, and SGR of sub-adult grass carp (P < 0.05). Appropriate levels of dietary leucine (11.91-17.92 g/kg) decreased the activities of serum parameters (glucose, cortisol, and methemoglobin contents, glutamic oxaloacetic transaminase, glutamic pyruvic transaminase, and lactate dehydrogenase), the contents of reactive oxygen species (ROS), nitric oxide (NO) and peroxynitrite (ONOO-). In addition, appropriate levels of dietary leucine (11.91-17.92 g/kg) increased the mRNA levels of mitochondrial biogenesis genes (PGC-1α, Nrf1/2, TFAM), fusion-related genes (Opa1, Mfn1/2) (P < 0.05), and decreased the mRNA levels of caspase 3, caspase 8, caspase 9, fission-related gene (Drp1), mitophagy-related genes (Pink1, Parkin) and autophagy-related genes (Beclin1, Ulk1, Atg5, Atg7, Atg12) (P < 0.05). Appropriate levels of dietary leucine (8.92-17.92 g/kg) also increased the protein levels of AMP-activated protein kinase (AMPK), prostacyclin (p62) and decreased the protein levels of protein light chain 3 (LC3), E3 ubiquitin ligase (Parkin), and Cytochrome c (Cytc). Appropriate levels of leucine (8.92-17.92 g/kg) could promote growth performance and alleviate nitrite-induced mitochondrial dysfunction, autophagy, apoptosis for sub-adult grass carp. Based on quadratic regression analysis of PWG and serum GPT activity, dietary leucine requirements of sub-adult grass carp were recommended to be 12.47 g/kg diet and 12.55 g/kg diet, respectively.
Collapse
Affiliation(s)
- Lu-Lu Zhen
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, Sichuan, China
| | - Shu-Wei Li
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, Sichuan, China
| | - Cheng-Bo Zhong
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, Sichuan, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China.
| |
Collapse
|
5
|
Forte M, D'Ambrosio L, Schiattarella GG, Salerno N, Perrone MA, Loffredo FS, Bertero E, Pilichou K, Manno G, Valenti V, Spadafora L, Bernardi M, Simeone B, Sarto G, Frati G, Perrino C, Sciarretta S. Mitophagy modulation for the treatment of cardiovascular diseases. Eur J Clin Invest 2024; 54:e14199. [PMID: 38530070 DOI: 10.1111/eci.14199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Defects of mitophagy, the selective form of autophagy for mitochondria, are commonly observed in several cardiovascular diseases and represent the main cause of mitochondrial dysfunction. For this reason, mitophagy has emerged as a novel and potential therapeutic target. METHODS In this review, we discuss current evidence about the biological significance of mitophagy in relevant preclinical models of cardiac and vascular diseases, such as heart failure, ischemia/reperfusion injury, metabolic cardiomyopathy and atherosclerosis. RESULTS Multiple studies have shown that cardiac and vascular mitophagy is an adaptive mechanism in response to stress, contributing to cardiovascular homeostasis. Mitophagy defects lead to cell death, ultimately impairing cardiac and vascular function, whereas restoration of mitophagy by specific compounds delays disease progression. CONCLUSIONS Despite previous efforts, the molecular mechanisms underlying mitophagy activation in response to stress are not fully characterized. A comprehensive understanding of different forms of mitophagy active in the cardiovascular system is extremely important for the development of new drugs targeting this process. Human studies evaluating mitophagy abnormalities in patients at high cardiovascular risk also represent a future challenge.
Collapse
Affiliation(s)
| | - Luca D'Ambrosio
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Gabriele G Schiattarella
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Nadia Salerno
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Marco Alfonso Perrone
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
- Clinical Pathways and Epidemiology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Francesco S Loffredo
- Division of Cardiology, Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Edoardo Bertero
- Department of Internal Medicine, University of Genova, Genoa, Italy
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino-Italian IRCCS Cardiology Network, Genoa, Italy
| | - Kalliopi Pilichou
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Girolamo Manno
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Valentina Valenti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- ICOT Istituto Marco Pasquali, Latina, Italy
| | | | - Marco Bernardi
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | | | | | - Giacomo Frati
- IRCCS Neuromed, Pozzilli, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Cinzia Perrino
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Sebastiano Sciarretta
- IRCCS Neuromed, Pozzilli, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
6
|
Wang Y, Xu T, Wang H, Xia G, Huang X. Inhibition of autophagy induced by tetrandrine promotes the accumulation of reactive oxygen species and sensitizes efficacy of tetrandrine in pancreatic cancer. Cancer Cell Int 2024; 24:241. [PMID: 38987818 PMCID: PMC11238362 DOI: 10.1186/s12935-024-03410-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/19/2024] [Indexed: 07/12/2024] Open
Abstract
Pancreatic cancer, characterized by its poor prognosis, exhibits a marked resistance to conventional chemotherapy and immunotherapy, underscoring the urgent need for more effective treatment modalities. In light of this, the present study is designed to assess the potential antineoplastic efficacy of a combined regimen involving tetrandrine, a plant-derived alkaloid, and autophagy inhibitors in the context of pancreatic cancer. Electron microscopy and immunoblots showed that tetrandrine promoted the formation of autophagosomes and the upregulation of LC3II and the downregulation of p62 expression, indicating that tetrandrine induced autophagy in pancreatic cancer cells. Western blot revealed that tetrandrine inhibited the phosphorylation of AKT and mTOR, as well as the expression of Bcl-2, while upregulating Beclin-1 expression. Moreover, tetrandrine promoted the transcription and protein expression of ATG7. Following the combination of autophagy inhibitors and tetrandrine, the apoptotic rate and cell death significantly increased in pancreatic cancer cells. Consistent results were obtained when ATG7 was silenced. Additionally, tetrandrine induced the generation of ROS, which was involved in the activation of autophagy and apoptosis. Further investigation revealed that upon autophagy inhibition, ROS accumulated in pancreatic cancer cells, resulting in decreased mitochondrial membrane potential and further induction of apoptosis. The results of treating subcutaneous xenograft tumors with a combination of tetrandrine and chloroquine validated that autophagy inhibition enhances the toxicity of tetrandrine against pancreatic cancer in vivo. Altogether, our study demonstrates that tetrandrine induces cytoprotective autophagy in pancreatic cancer cells. Inhibiting tetrandrine-induced autophagy promotes the accumulation of ROS and enhances its toxicity against pancreatic cancer.
Collapse
Affiliation(s)
- Yiwei Wang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, China
| | - Ting Xu
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, China
| | - Hongcheng Wang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, China
| | - Guanggai Xia
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, China.
| | - Xinyu Huang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Rd, Shanghai, 200233, China.
| |
Collapse
|
7
|
Liu BH, Xu CZ, Liu Y, Lu ZL, Fu TL, Li GR, Deng Y, Luo GQ, Ding S, Li N, Geng Q. Mitochondrial quality control in human health and disease. Mil Med Res 2024; 11:32. [PMID: 38812059 PMCID: PMC11134732 DOI: 10.1186/s40779-024-00536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.
Collapse
Affiliation(s)
- Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Qing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
8
|
Liu M, He L. Dietary cysteine and methionine promote peroxisome elevation and fat loss by induction of CG33474 expression in Drosophila adipose tissue. Cell Mol Life Sci 2024; 81:190. [PMID: 38649521 PMCID: PMC11035426 DOI: 10.1007/s00018-024-05226-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/13/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024]
Abstract
The high-protein diet (HPD) has emerged as a potent dietary approach to curb obesity. Peroxisome, a highly malleable organelle, adapts to nutritional changes to maintain homeostasis by remodeling its structure, composition, and quantity. However, the impact of HPD on peroxisomes and the underlying mechanism remains elusive. Using Drosophila melanogaster as a model system, we discovered that HPD specifically increases peroxisome levels within the adipose tissues. This HPD-induced peroxisome elevation is attributed to cysteine and methionine by triggering the expression of CG33474, a fly homolog of mammalian PEX11G. Both the overexpression of Drosophila CG33474 and human PEX11G result in increased peroxisome size. In addition, cysteine and methionine diets both reduce lipid contents, a process that depends on the presence of CG33474. Furthermore, CG33474 stimulates the breakdown of neutral lipids in a cell-autonomous manner. Moreover, the expression of CG33474 triggered by cysteine and methionine requires TOR signaling. Finally, we found that CG33474 promotes inter-organelle contacts between peroxisomes and lipid droplets (LDs), which might be a potential mechanism for CG33474-induced fat loss. In summary, our findings demonstrate that CG33474/PEX11G may serve as an essential molecular bridge linking HPD to peroxisome dynamics and lipid metabolism.
Collapse
Affiliation(s)
- Meng Liu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Li He
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
9
|
Ben Dhaou C, Scott ML, Orr AW. Advances in Understanding Cardiovascular Disease Pathogenesis through Next-Generation Technologies. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:476-481. [PMID: 38519246 PMCID: PMC10988757 DOI: 10.1016/j.ajpath.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 03/24/2024]
Affiliation(s)
- Cyrine Ben Dhaou
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, Louisiana
| | - Matthew L Scott
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, Louisiana
| | - A Wayne Orr
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, Shreveport, Louisiana; Department of Molecular and Cellular Physiology, LSU Health Shreveport, Shreveport, Louisiana; Department of Cell Biology and Anatomy, LSU Health Shreveport, Shreveport, Louisiana.
| |
Collapse
|
10
|
Li SY, Lu ZH, Leung JCS, Kwok TCY. Association of dietary protein intake, inflammation with muscle mass, physical performance and incident sarcopenia in Chinese community-dwelling older adults. J Nutr Health Aging 2024; 28:100163. [PMID: 38350300 DOI: 10.1016/j.jnha.2024.100163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/08/2023] [Indexed: 02/15/2024]
Abstract
OBJECTIVES Inflammation and impaired muscle synthesis are important factors of sarcopenia. Plant protein may reduce inflammation but may not be as efficient as animal protein in providing essential amino acids. We therefore examined the associations between dietary protein intake and changes in muscle mass and physical performance, incident sarcopenia, and the interaction effect of inflammation. DESIGN Prospective cohort study. SETTING The Mr. OS and Ms. OS (Hong Kong) cohort. PARTICIPANTS A total of 2,811 sarcopenia-free participants and 569 sarcopenia participants aged ≥65 years were recruited from communities. MEASUREMENTS Dietary protein intake was assessed using a validated food frequency questionnaire. Serum high-sensitivity C-reactive protein (hs-CRP) was measured. Linear regression examined the associations between dietary protein intake and 4-year changes in muscle mass and physical performance. Cox regression examined the association between dietary protein intake and incident sarcopenia. RESULTS Higher plant protein intake, but not total and animal protein, was associated with less decline in muscle mass and gait speed among sarcopenia-free participants. Conversely, higher ratio of animal-to-plant protein was associated with reduced muscle mass loss among participants with sarcopenia. The highest tertile of plant protein intake was associated with lower incident sarcopenia risk (HR: 0.75, 95% CI: 0.57-0.98; P-trend = 0.034) compared to the lowest tertile. Notably, this association was observed among participants with higher serum hs-CRP levels (HR: 0.57, 95% CI: 0.34-0.95), but not in those with lower hs-CRP levels. CONCLUSION Dietary animal and plant protein intake have differential associations with muscle mass and physical performance in older adults with and without sarcopenia. The role of plant protein in preventing sarcopenia involves modulation of inflammation.
Collapse
Affiliation(s)
- Shu-Yi Li
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhi-Hui Lu
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jason C S Leung
- Jockey Club Centre for Osteoporosis Care and Control, The Chinese University of Hong Kong, Hong Kong, China
| | - Timothy C Y Kwok
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China; Jockey Club Centre for Osteoporosis Care and Control, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
11
|
Zimmermann A, Madeo F, Diwan A, Sadoshima J, Sedej S, Kroemer G, Abdellatif M. Metabolic control of mitophagy. Eur J Clin Invest 2024; 54:e14138. [PMID: 38041247 DOI: 10.1111/eci.14138] [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: 10/03/2023] [Revised: 11/09/2023] [Accepted: 11/20/2023] [Indexed: 12/03/2023]
Abstract
Mitochondrial dysfunction is a major hallmark of ageing and related chronic disorders. Controlled removal of damaged mitochondria by the autophagic machinery, a process known as mitophagy, is vital for mitochondrial homeostasis and cell survival. The central role of mitochondria in cellular metabolism places mitochondrial removal at the interface of key metabolic pathways affecting the biosynthesis or catabolism of acetyl-coenzyme A, nicotinamide adenine dinucleotide, polyamines, as well as fatty acids and amino acids. Molecular switches that integrate the metabolic status of the cell, like AMP-dependent protein kinase, protein kinase A, mechanistic target of rapamycin and sirtuins, have also emerged as important regulators of mitophagy. In this review, we discuss how metabolic regulation intersects with mitophagy. We place special emphasis on the metabolic regulatory circuits that may be therapeutically targeted to delay ageing and mitochondria-associated chronic diseases. Moreover, we identify outstanding knowledge gaps, such as the ill-defined distinction between basal and damage-induced mitophagy, which must be resolved to boost progress in this area.
Collapse
Affiliation(s)
- Andreas Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth-University of Graz, Graz, Austria
| | - Frank Madeo
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth-University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Abhinav Diwan
- Division of Cardiology and Center for Cardiovascular Research, Washington University School of Medicine, and John Cochran Veterans Affairs Medical Center, St. Louis, Missouri, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Simon Sedej
- BioTechMed Graz, Graz, Austria
- Department of Cardiology, Medical University of Graz, Graz, Austria
- Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Department of Biology, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, Paris, France
| | - Mahmoud Abdellatif
- BioTechMed Graz, Graz, Austria
- Department of Cardiology, Medical University of Graz, Graz, Austria
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
| |
Collapse
|
12
|
Anand SK, Governale TA, Zhang X, Razani B, Yurdagul A, Pattillo CB, Rom O. Amino Acid Metabolism and Atherosclerotic Cardiovascular Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:510-524. [PMID: 38171450 PMCID: PMC10988767 DOI: 10.1016/j.ajpath.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/09/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024]
Abstract
Despite significant advances in medical treatments and drug development, atherosclerotic cardiovascular disease (ASCVD) remains a leading cause of death worldwide. Dysregulated lipid metabolism is a well-established driver of ASCVD. Unfortunately, even with potent lipid-lowering therapies, ASCVD-related deaths have continued to increase over the past decade, highlighting an incomplete understanding of the underlying risk factors and mechanisms of ASCVD. Accumulating evidence over the past decades indicates a correlation between amino acids and disease state. This review explores the emerging role of amino acid metabolism in ASCVD, uncovering novel potential biomarkers, causative factors, and therapeutic targets. Specifically, the significance of arginine and its related metabolites, homoarginine and polyamines, branched-chain amino acids, glycine, and aromatic amino acids, in ASCVD are discussed. These amino acids and their metabolites have been implicated in various processes characteristic of ASCVD, including impaired lipid metabolism, endothelial dysfunction, increased inflammatory response, and necrotic core development. Understanding the complex interplay between dysregulated amino acid metabolism and ASCVD provides new insights that may lead to the development of novel diagnostic and therapeutic approaches. Although further research is needed to uncover the precise mechanisms involved, it is evident that amino acid metabolism plays a role in ASCVD.
Collapse
Affiliation(s)
- Sumit Kumar Anand
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Theresea-Anne Governale
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Xiangyu Zhang
- Division of Cardiology and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Babak Razani
- Division of Cardiology and Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Arif Yurdagul
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Christopher B Pattillo
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana.
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana.
| |
Collapse
|
13
|
Stroope C, Nettersheim FS, Coon B, Finney AC, Schwartz MA, Ley K, Rom O, Yurdagul A. Dysregulated cellular metabolism in atherosclerosis: mediators and therapeutic opportunities. Nat Metab 2024; 6:617-638. [PMID: 38532071 PMCID: PMC11055680 DOI: 10.1038/s42255-024-01015-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 02/20/2024] [Indexed: 03/28/2024]
Abstract
Accumulating evidence over the past decades has revealed an intricate relationship between dysregulation of cellular metabolism and the progression of atherosclerotic cardiovascular disease. However, an integrated understanding of dysregulated cellular metabolism in atherosclerotic cardiovascular disease and its potential value as a therapeutic target is missing. In this Review, we (1) summarize recent advances concerning the role of metabolic dysregulation during atherosclerosis progression in lesional cells, including endothelial cells, vascular smooth muscle cells, macrophages and T cells; (2) explore the complexity of metabolic cross-talk between these lesional cells; (3) highlight emerging technologies that promise to illuminate unknown aspects of metabolism in atherosclerosis; and (4) suggest strategies for targeting these underexplored metabolic alterations to mitigate atherosclerosis progression and stabilize rupture-prone atheromas with a potential new generation of cardiovascular therapeutics.
Collapse
Affiliation(s)
- Chad Stroope
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Felix Sebastian Nettersheim
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Brian Coon
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Cardiovascular Biology Research Program, OMRF, Oklahoma City, OK, USA
- Department of Cell Biology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - Alexandra C Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
- Immunology Center of Georgia (IMMCG), Augusta University Immunology Center of Georgia, Augusta, GA, USA
| | - Oren Rom
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
14
|
Huang N, Winans T, Wyman B, Oaks Z, Faludi T, Choudhary G, Lai ZW, Lewis J, Beckford M, Duarte M, Krakko D, Patel A, Park J, Caza T, Sadeghzadeh M, Morel L, Haas M, Middleton F, Banki K, Perl A. Rab4A-directed endosome traffic shapes pro-inflammatory mitochondrial metabolism in T cells via mitophagy, CD98 expression, and kynurenine-sensitive mTOR activation. Nat Commun 2024; 15:2598. [PMID: 38519468 PMCID: PMC10960037 DOI: 10.1038/s41467-024-46441-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 02/28/2024] [Indexed: 03/25/2024] Open
Abstract
Activation of the mechanistic target of rapamycin (mTOR) is a key metabolic checkpoint of pro-inflammatory T-cell development that contributes to the pathogenesis of autoimmune diseases, such as systemic lupus erythematosus (SLE), however, the underlying mechanisms remain poorly understood. Here, we identify a functional role for Rab4A-directed endosome traffic in CD98 receptor recycling, mTOR activation, and accumulation of mitochondria that connect metabolic pathways with immune cell lineage development and lupus pathogenesis. Based on integrated analyses of gene expression, receptor traffic, and stable isotope tracing of metabolic pathways, constitutively active Rab4AQ72L exerts cell type-specific control over metabolic networks, dominantly impacting CD98-dependent kynurenine production, mTOR activation, mitochondrial electron transport and flux through the tricarboxylic acid cycle and thus expands CD4+ and CD3+CD4-CD8- double-negative T cells over CD8+ T cells, enhancing B cell activation, plasma cell development, antinuclear and antiphospholipid autoantibody production, and glomerulonephritis in lupus-prone mice. Rab4A deletion in T cells and pharmacological mTOR blockade restrain CD98 expression, mitochondrial metabolism and lineage skewing and attenuate glomerulonephritis. This study identifies Rab4A-directed endosome traffic as a multilevel regulator of T cell lineage specification during lupus pathogenesis.
Collapse
Affiliation(s)
- Nick Huang
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Thomas Winans
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Brandon Wyman
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Zachary Oaks
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Tamas Faludi
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Gourav Choudhary
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Zhi-Wei Lai
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Joshua Lewis
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Miguel Beckford
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Manuel Duarte
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Daniel Krakko
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Akshay Patel
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Joy Park
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Tiffany Caza
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Mahsa Sadeghzadeh
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Mark Haas
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Frank Middleton
- Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Katalin Banki
- Department of Pathology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA
| | - Andras Perl
- Department of Medicine, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA.
- Department of Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA.
- Department of Microbiology and Immunology, State University of New York, Upstate Medical University, Norton College of Medicine, Syracuse, New York, NY, 13210, USA.
| |
Collapse
|
15
|
Yan H, Liu Y, Li X, Yu B, He J, Mao X, Yu J, Huang Z, Luo Y, Luo J, Wu A, Chen D. Leucine alleviates cytokine storm syndrome by regulating macrophage polarization via the mTORC1/LXRα signaling pathway. eLife 2024; 12:RP89750. [PMID: 38442142 PMCID: PMC10942637 DOI: 10.7554/elife.89750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Abstract
Cytokine storms are associated with severe pathological damage and death in some diseases. Excessive activation of M1 macrophages and the subsequent secretion of pro-inflammatory cytokines are a major cause of cytokine storms. Therefore, promoting the polarization of M2 macrophages to restore immune balance is a promising therapeutic strategy for treating cytokine storm syndrome (CSS). This study was aimed at investigating the potential protective effects of leucine on lipopolysaccharide (LPS)-induced CSS in mice and exploring the underlying mechanisms. CSS was induced by LPS administration in mice, which were concurrently administered leucine orally. In vitro, bone marrow derived macrophages (BMDMs) were polarized to M1 and M2 phenotypes with LPS and interleukin-4 (IL-4), respectively, and treated with leucine. Leucine decreased mortality in mice treated with lethal doses of LPS. Specifically, leucine decreased M1 polarization and promoted M2 polarization, thus diminishing pro-inflammatory cytokine levels and ameliorating CSS in mice. Further studies revealed that leucine-induced macrophage polarization through the mechanistic target of rapamycin complex 1 (mTORC1)/liver X receptor α (LXRα) pathway, which synergistically enhanced the expression of the IL-4-induced M2 marker Arg1 and subsequent M2 polarization. In summary, this study revealed that leucine ameliorates CSS in LPS mice by promoting M2 polarization through the mTORC1/LXRα/Arg1 signaling pathway. Our findings indicate that a fundamental link between metabolism and immunity contributes to the resolution of inflammation and the repair of damaged tissues.
Collapse
Affiliation(s)
- Hui Yan
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Yao Liu
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Xipeng Li
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Bing Yu
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Jun He
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Xiangbing Mao
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Jie Yu
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Zhiqing Huang
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Yuheng Luo
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Junqiu Luo
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Aimin Wu
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| | - Daiwen Chen
- Key Laboratory of Animal Disease Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease resistant Nutrition of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural UniversityChengduChina
| |
Collapse
|
16
|
Rose AJ, Rusu PM. A leucine-macrophage mTORC1 connection drives increased risk of atherosclerosis with high-protein diets. Nat Metab 2024; 6:203-204. [PMID: 38409322 DOI: 10.1038/s42255-023-00952-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Affiliation(s)
- Adam J Rose
- Nutrient Metabolism & Signalling Laboratory, Dept. Of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia.
| | - Patricia M Rusu
- Nutrient Metabolism & Signalling Laboratory, Dept. Of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
17
|
Tyagi SC, Pushpakumar S, Sen U, Akinterinwa OE, Zheng Y, Mokshagundam SPL, Kalra DK, Singh M. Role of circadian clock system in the mitochondrial trans-sulfuration pathway and tissue remodeling. Can J Physiol Pharmacol 2024; 102:105-115. [PMID: 37979203 DOI: 10.1139/cjpp-2023-0186] [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: 11/20/2023]
Abstract
Previous studies from our laboratory revealed that the gaseous molecule hydrogen sulfide (H2S), a metabolic product of epigenetics, involves trans-sulfuration pathway for ensuring metabolism and clearance of homocysteine (Hcy) from body, thereby mitigating the skeletal muscle's pathological remodeling. Although the master circadian clock regulator that is known as brain and muscle aryl hydrocarbon receptor nuclear translocator like protein 1 (i.e., BMAL 1) is associated with S-adenosylhomocysteine hydrolase (SAHH) and Hcy metabolism but how trans-sulfuration pathway is influenced by the circadian clock remains unexplored. We hypothesize that alterations in the functioning of circadian clock during sleep and wake cycle affect skeletal muscle's biology. To test this hypothesis, we measured serum matrix metalloproteinase (MMP) activities using gelatin gels for analyzing the MMP-2 and MMP-9. Further, employing casein gels, we also studied MMP-13 that is known to be influenced by the growth arrest and DNA damage-45 (GADD45) protein during sleep and wake cycle. The wild type and cystathionine β synthase-deficient (CBS-/+) mice strains were treated with H2S and subjected to measurement of trans-sulfuration factors from skeletal muscle tissues. The results suggested highly robust activation of MMPs in the wake mice versus sleep mice, which appears somewhat akin to the "1-carbon metabolic dysregulation", which takes place during remodeling of extracellular matrix during muscular dystrophy. Interestingly, the levels of trans-sulfuration factors such as CBS, cystathionine γ lyase (CSE), methyl tetrahydrofolate reductase (MTHFR), phosphatidylethanolamine N-methyltransferase (PEMT), and Hcy-protein bound paraoxonase 1 (PON1) were attenuated in CBS-/+ mice. However, treatment with H2S mitigated the attenuation of the trans-sulfuration pathway. In addition, levels of mitochondrial peroxisome proliferator-activated receptor-gamma coactivator 1-α (PGC 1-α) and mitofusin-2 (MFN-2) were significantly improved by H2S intervention. Our findings suggest participation of the circadian clock in trans-sulfuration pathway that affects skeletal muscle remodeling and mitochondrial regeneration.
Collapse
Affiliation(s)
- Suresh C Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Sathnur Pushpakumar
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Utpal Sen
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Oluwaseun E Akinterinwa
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Yuting Zheng
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Sri Prakash L Mokshagundam
- Division of Endocrinology, Metabolism and Diabetes and Robley Rex VA Medical Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Dinesh K Kalra
- Division of Cardiovascular Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Mahavir Singh
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
18
|
Zhang X, Kapoor D, Jeong SJ, Fappi A, Stitham J, Shabrish V, Sergin I, Yousif E, Rodriguez-Velez A, Yeh YS, Park A, Yurdagul A, Rom O, Epelman S, Schilling JD, Sardiello M, Diwan A, Cho J, Stitziel NO, Javaheri A, Lodhi IJ, Mittendorfer B, Razani B. Identification of a leucine-mediated threshold effect governing macrophage mTOR signalling and cardiovascular risk. Nat Metab 2024; 6:359-377. [PMID: 38409323 PMCID: PMC11448845 DOI: 10.1038/s42255-024-00984-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 01/09/2024] [Indexed: 02/28/2024]
Abstract
High protein intake is common in western societies and is often promoted as part of a healthy lifestyle; however, amino-acid-mediated mammalian target of rapamycin (mTOR) signalling in macrophages has been implicated in the pathogenesis of ischaemic cardiovascular disease. In a series of clinical studies on male and female participants ( NCT03946774 and NCT03994367 ) that involved graded amounts of protein ingestion together with detailed plasma amino acid analysis and human monocyte/macrophage experiments, we identify leucine as the key activator of mTOR signalling in macrophages. We describe a threshold effect of high protein intake and circulating leucine on monocytes/macrophages wherein only protein in excess of ∼25 g per meal induces mTOR activation and functional effects. By designing specific diets modified in protein and leucine content representative of the intake in the general population, we confirm this threshold effect in mouse models and find ingestion of protein in excess of ∼22% of dietary energy requirements drives atherosclerosis in male mice. These data demonstrate a mechanistic basis for the adverse impact of excessive dietary protein on cardiovascular risk.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Divya Kapoor
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St Louis, MO, USA
| | - Se-Jin Jeong
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Alan Fappi
- Division of Nutritional Science and Obesity Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Departments of Medicine and Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Jeremiah Stitham
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | - Vasavi Shabrish
- Division of Nutritional Science and Obesity Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Departments of Medicine and Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Ismail Sergin
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Eman Yousif
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | | | - Yu-Sheng Yeh
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Arick Park
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Oren Rom
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Slava Epelman
- Peter Munk Cardiac Center and University Health Network, University of Toronto, Toronto, Canada
| | - Joel D Schilling
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Marco Sardiello
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Abhinav Diwan
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St Louis, MO, USA
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Nathan O Stitziel
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Ali Javaheri
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St Louis, MO, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | - Bettina Mittendorfer
- Division of Nutritional Science and Obesity Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Departments of Medicine and Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA.
| | - Babak Razani
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA.
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
19
|
Sun WX, Zhang KH, Zhou Q, Hu SH, Lin Y, Xu W, Zhao SM, Yuan YY. Tryptophanylation of insulin receptor by WARS attenuates insulin signaling. Cell Mol Life Sci 2024; 81:25. [PMID: 38212570 PMCID: PMC11072365 DOI: 10.1007/s00018-023-05082-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 01/13/2024]
Abstract
Increased circulating amino acid levels have been linked to insulin resistance and development of type 2 diabetes (T2D), but the underlying mechanism remains largely unknown. Herein, we show that tryptophan modifies insulin receptor (IR) to attenuate insulin signaling and impair glucose uptake. Mice fed with tryptophan-rich chow developed insulin resistance. Excessive tryptophan promoted tryptophanyl-tRNA synthetase (WARS) to tryptophanylate lysine 1209 of IR (W-K1209), which induced insulin resistance by inhibiting the insulin-stimulated phosphorylation of IR, AKT, and AS160. SIRT1, but not other sirtuins, detryptophanylated IRW-K1209 to increase the insulin sensitivity. Collectively, we unveiled the mechanisms of how tryptophan impaired insulin signaling, and our data suggested that WARS might be a target to attenuate insulin resistance in T2D patients.
Collapse
Affiliation(s)
- Wen-Xing Sun
- Obstetrics and Gynecology Hospital of Fudan University, Institutes of Biomedical Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, People's Republic of China
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, People's Republic of China
| | - Kai-Hui Zhang
- Obstetrics and Gynecology Hospital of Fudan University, Institutes of Biomedical Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, People's Republic of China
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Jinan, People's Republic of China
- Children's Research Institute, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), Jinan, People's Republic of China
| | - Qian Zhou
- Obstetrics and Gynecology Hospital of Fudan University, Institutes of Biomedical Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, People's Republic of China
- NHC Key Lab of Reproduction Regulation, Shanghai Key Laboratory of Metabolic Remodeling and Health, and Children's Hospital of Fudan University, Shanghai, People's Republic of China
| | - Song-Hua Hu
- Obstetrics and Gynecology Hospital of Fudan University, Institutes of Biomedical Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, People's Republic of China
- NHC Key Lab of Reproduction Regulation, Shanghai Key Laboratory of Metabolic Remodeling and Health, and Children's Hospital of Fudan University, Shanghai, People's Republic of China
| | - Yan Lin
- Obstetrics and Gynecology Hospital of Fudan University, Institutes of Biomedical Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, People's Republic of China
- NHC Key Lab of Reproduction Regulation, Shanghai Key Laboratory of Metabolic Remodeling and Health, and Children's Hospital of Fudan University, Shanghai, People's Republic of China
- Shanghai Fifth People's Hospital of Fudan University, Fudan University, Shanghai, People's Republic of China
| | - Wei Xu
- Obstetrics and Gynecology Hospital of Fudan University, Institutes of Biomedical Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, People's Republic of China
- NHC Key Lab of Reproduction Regulation, Shanghai Key Laboratory of Metabolic Remodeling and Health, and Children's Hospital of Fudan University, Shanghai, People's Republic of China
- Shanghai Fifth People's Hospital of Fudan University, Fudan University, Shanghai, People's Republic of China
| | - Shi-Min Zhao
- Obstetrics and Gynecology Hospital of Fudan University, Institutes of Biomedical Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, People's Republic of China.
- NHC Key Lab of Reproduction Regulation, Shanghai Key Laboratory of Metabolic Remodeling and Health, and Children's Hospital of Fudan University, Shanghai, People's Republic of China.
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, College of Pharmacy, Qinghai University for Nationalities, Xining, People's Republic of China.
| | - Yi-Yuan Yuan
- Obstetrics and Gynecology Hospital of Fudan University, Institutes of Biomedical Sciences, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, People's Republic of China.
- NHC Key Lab of Reproduction Regulation, Shanghai Key Laboratory of Metabolic Remodeling and Health, and Children's Hospital of Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
20
|
Song J, Wu J, Robichaux DJ, Li T, Wang S, Arredondo Sancristobal MJ, Dong B, Dobrev D, Karch J, Thomas SS, Li N. A High-Protein Diet Promotes Atrial Arrhythmogenesis via Absent-in-Melanoma 2 Inflammasome. Cells 2024; 13:108. [PMID: 38247800 PMCID: PMC10814244 DOI: 10.3390/cells13020108] [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: 12/17/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
High-protein diets (HPDs) offer health benefits, such as weight management and improved metabolic profiles. The effects of HPD on cardiac arrhythmogenesis remain unclear. Atrial fibrillation (AF), the most common arrhythmia, is associated with inflammasome activation. The role of the Absent-in-Melanoma 2 (AIM2) inflammasome in AF pathogenesis remains unexplored. In this study, we discovered that HPD increased susceptibility to AF. To demonstrate the involvement of AIM2 signaling in the pathogenesis of HPD-induced AF, wildtype (WT) and Aim2-/- mice were fed normal-chow (NC) and HPD, respectively. Four weeks later, inflammasome activity was upregulated in the atria of WT-HPD mice, but not in the Aim2-/--HPD mice. The increased AF vulnerability in WT-HPD mice was associated with abnormal sarcoplasmic reticulum (SR) Ca2+-release events in atrial myocytes. HPD increased the cytoplasmic double-strand (ds) DNA level, causing AIM2 activation. Genetic inhibition of AIM2 in Aim2-/- mice reduced susceptibility to AF, cytoplasmic dsDNA level, mitochondrial ROS production, and abnormal SR Ca2+-release in atrial myocytes. These data suggest that HPD creates a substrate conducive to AF development by activating the AIM2-inflammasome, which is associated with mitochondrial oxidative stress along with proarrhythmic SR Ca2+-release. Our data imply that targeting the AIM2 inflammasome might constitute a novel anti-AF strategy in certain patient subpopulations.
Collapse
Affiliation(s)
- Jia Song
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX 77030, USA (M.J.A.S.)
| | - Jiao Wu
- Department of Medicine, Section of Nephrology, Houston, TX 77030, USA
| | - Dexter J. Robichaux
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA (D.D.)
| | - Tingting Li
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX 77030, USA (M.J.A.S.)
| | - Shuyue Wang
- Department of Medicine, Section of Gastroenterology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Bingning Dong
- Department of Medicine, Section of Gastroenterology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dobromir Dobrev
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA (D.D.)
- Institute of Pharmacology, University Duisburg-Essen, 45147 Essen, Germany
- Department of Medicine, Montreal Heart Institute, Université de Montréal, Montréal, QC H1T 1C8, Canada
| | - Jason Karch
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA (D.D.)
| | - Sandhya S. Thomas
- Department of Medicine, Section of Nephrology, Houston, TX 77030, USA
- Michael E. Debakey VA Medical Center, Houston, TX 77030, USA
| | - Na Li
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX 77030, USA (M.J.A.S.)
| |
Collapse
|
21
|
Pan W, Zhang J, Zhang L, Zhang Y, Song Y, Han L, Tan M, Yin Y, Yang T, Jiang T, Li H. Comprehensive view of macrophage autophagy and its application in cardiovascular diseases. Cell Prolif 2024; 57:e13525. [PMID: 37434325 PMCID: PMC10771119 DOI: 10.1111/cpr.13525] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 07/13/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the primary drivers of the growing public health epidemic and the leading cause of premature mortality and economic burden worldwide. With decades of research, CVDs have been proven to be associated with the dysregulation of the inflammatory response, with macrophages playing imperative roles in influencing the prognosis of CVDs. Autophagy is a conserved pathway that maintains cellular functions. Emerging evidence has revealed an intrinsic connection between autophagy and macrophage functions. This review focuses on the role and underlying mechanisms of autophagy-mediated regulation of macrophage plasticity in polarization, inflammasome activation, cytokine secretion, metabolism, phagocytosis, and the number of macrophages. In addition, autophagy has been shown to connect macrophages and heart cells. It is attributed to specific substrate degradation or signalling pathway activation by autophagy-related proteins. Referring to the latest reports, applications targeting macrophage autophagy have been discussed in CVDs, such as atherosclerosis, myocardial infarction, heart failure, and myocarditis. This review describes a novel approach for future CVD therapies.
Collapse
Affiliation(s)
- Wanqian Pan
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Jun Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Lei Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yue Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yiyi Song
- Suzhou Medical College of Soochow UniversitySuzhouChina
| | - Lianhua Han
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Mingyue Tan
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yunfei Yin
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Tianke Yang
- Department of Ophthalmology, Eye Institute, Eye & ENT HospitalFudan UniversityShanghaiChina
- Department of OphthalmologyThe First Affiliated Hospital of USTC, University of Science and Technology of ChinaHefeiChina
| | - Tingbo Jiang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Hongxia Li
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
22
|
Zeng GG, Tang SS, Jiang WL, Yu J, Nie GY, Tang CK. Apelin-13: A Protective Role in Vascular Diseases. Curr Probl Cardiol 2024; 49:102088. [PMID: 37716542 DOI: 10.1016/j.cpcardiol.2023.102088] [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: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023]
Abstract
Vascular disease is a common problem with high mortality all over the world. Apelin-13, a key subtype of apelin, takes part in many physiological and pathological responses via regulating many target genes and target molecules or participating in many signaling pathways. More and more studies have demonstrated that apelin-13 is implicated in the onset and progression of vascular disease in recent years. It has been shown that apelin-13 could ameliorate vascular disease by inhibiting inflammation, restraining apoptosis, suppressing oxidative stress, and facilitating autophagy. In this article, we sum up the progress of apelin-13 in the occurrence and development of vascular disease and offer some insightful views about the treatment and prevention strategies of vascular disease.
Collapse
Affiliation(s)
- Guang-Gui Zeng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan, China; The Seventh Affiliated Hospital University of South China/ Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, People's Republic of China
| | - Shang-Shu Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan, China; The Seventh Affiliated Hospital University of South China/ Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, People's Republic of China
| | - Wan-Li Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan, China; The Seventh Affiliated Hospital University of South China/ Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, People's Republic of China
| | - Jiang Yu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan, China; The Seventh Affiliated Hospital University of South China/ Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, People's Republic of China
| | - Gui-Ying Nie
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan, China; The Seventh Affiliated Hospital University of South China/ Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, People's Republic of China
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan, China; The Seventh Affiliated Hospital University of South China/ Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, People's Republic of China.
| |
Collapse
|
23
|
Schwartz BH, Choi SY, Mathews A, Aggarwal M, Handberg EM, Pepine CJ, Rogers W, Reis S, Cook-Wiens G, Merz CNB, Wei J. Dietary Composition, Angiographic Coronary Disease, and Cardiovascular Outcomes in the WISE Study (Women's Ischemia Syndrome Evaluation). J Clin Med 2023; 12:7762. [PMID: 38137831 PMCID: PMC10743715 DOI: 10.3390/jcm12247762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Studies relating diet to angiographic coronary artery disease (CAD) and subsequent major adverse cardiac events (MACE) in women are limited. Information on diet was collected in the Women's Ischemia Syndrome Evaluation (WISE), a prospective cohort study of symptomatic women referred for coronary angiography to evaluate suspected ischemic heart disease. METHODS A consecutive subgroup (n = 201 of 936) of enrolled women completed the modified Block food frequency questionnaire (FFQ). Data on outcomes were collected and adjudicated after 8-year follow-up. A set of logistic regression models were fitted for non-obstructive versus obstructive coronary stenosis (<50% versus ≥50%). Cox proportional hazard regression models were fitted for outcomes, with each dietary composition variable adjusted for the degree of coronary stenosis. RESULTS At baseline, the subgroup cohort was 58 ± 12 years old with a body mass index (BMI) of 30 ± 7 kg/m2. An increased proportion of calories consumed from protein was associated with higher levels of baseline obstructive coronary stenosis. Those individuals who ate a higher amount of protein, carotene, and servings of vegetables and meat, however, were each associated with lower subsequent adverse outcomes, respectively. CONCLUSIONS Among women undergoing coronary angiography for suspected CAD, a higher percentage of protein intake was associated with higher baseline stenosis severity; however, the amount of protein intake, vegetable, meat, and carotene intake, was conversely associated with subsequent lower adverse cardiovascular outcome risk.
Collapse
Affiliation(s)
- Brandon H. Schwartz
- Barbra Streisand Women’s Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA 90048, USA; (B.H.S.); (C.N.B.M.)
| | - So Yung Choi
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (S.Y.C.); (G.C.-W.)
| | - Anne Mathews
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL 32611, USA; (A.M.); (M.A.); (E.M.H.)
| | - Monica Aggarwal
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL 32611, USA; (A.M.); (M.A.); (E.M.H.)
| | - Eileen M. Handberg
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL 32611, USA; (A.M.); (M.A.); (E.M.H.)
| | - Carl J. Pepine
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL 32611, USA; (A.M.); (M.A.); (E.M.H.)
| | - William Rogers
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Steven Reis
- Division of Health Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Galen Cook-Wiens
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (S.Y.C.); (G.C.-W.)
| | - C. Noel Bairey Merz
- Barbra Streisand Women’s Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA 90048, USA; (B.H.S.); (C.N.B.M.)
| | - Janet Wei
- Barbra Streisand Women’s Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA 90048, USA; (B.H.S.); (C.N.B.M.)
| |
Collapse
|
24
|
Tan B, Zheng X, Xie X, Chen Y, Li Y, He W. MMP11 and MMP14 contribute to the interaction between castration-resistant prostate cancer and adipocytes. Am J Cancer Res 2023; 13:5934-5949. [PMID: 38187060 PMCID: PMC10767328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/10/2023] [Indexed: 01/09/2024] Open
Abstract
Previous studies have demonstrated that adipocytes promote prostate cancer (PCa) cell progression, which facilitates the development of PCa into castration-resistant prostate cancer (CRPC); however, the underlying mechanisms are still not fully understood. Matrix metalloproteinases (MMPs) are a group of proteases responsible for the degradation of extracellular matrix (ECM) and the activation of latent factors. In our study, we detected that MMP11 expression was increased in PCa patients and that a high level of MMP11 was correlated with poor prognosis. Furthermore, siRNA knockdown of MMP11 in CRPC cells not only blocked the delipidation and dedifferentiation of mature adipocytes but also reduced the lipid uptake and utilization of CRPC cells in a cell co-culture model. The number of mitophagosomes and the expression level of Parkin were increased in MMP11-silenced CRPC cells. Moreover, we found that simultaneous downregulation of MMP14 and MMP11 expression may benefit patient survival. Indeed, MMP11/14 knockdown in CRPC cells significantly decreased lipid metabolism and cell invasion, at least partly through the mTOR/HIF1α/MMP2 signaling pathway. Importantly, MMP11/14 knockdown dramatically delayed tumor growth in a xenograft mouse model. Consistently, the decreased lipid metabolism, Ki67 and MMP2 expression, as well as the increased Parkin level were also confirmed in in vivo experiments, further demonstrating the mechanisms responsible for the tumor-promoting effects of MMP11/14. Collectively, our study elucidated the role of MMP11 and MMP14 in the bidirectional crosstalk between adipocytes and CRPC cells and provided the rationale of targeting MMP11/14 for the treatment of CRPC patients.
Collapse
Affiliation(s)
- Bing Tan
- Department of Urology, University-Town Hospital of Chongqing Medical UniversityShapingba District, Chongqing 401331, China
- Medical Sciences Research Center, University-Town Hospital of Chongqing Medical UniversityShapingba District, Chongqing 401331, China
- Department of Urology, The First Affiliated Hospital of Chongqing Medical UniversityYuzhong District, Chongqing 400016, China
| | - Xiaoyu Zheng
- School of Clinical Medicine, Chongqing Medical and Pharmaceutical CollegeShapingba District, Chongqing 401331, China
| | - Xiaoqin Xie
- Department of Clinical Laboratory, Chongqing Blood CenterJiulongpo District, Chongqing 400015, China
| | - Yirong Chen
- Department of Urology, University-Town Hospital of Chongqing Medical UniversityShapingba District, Chongqing 401331, China
| | - Yuehua Li
- Department of Urology, University-Town Hospital of Chongqing Medical UniversityShapingba District, Chongqing 401331, China
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical UniversityYuzhong District, Chongqing 400016, China
| |
Collapse
|
25
|
Kontush A, Martin M, Brites F. Sweet swell of burning fat: emerging role of high-density lipoprotein in energy homeostasis. Curr Opin Lipidol 2023; 34:235-242. [PMID: 37797204 DOI: 10.1097/mol.0000000000000904] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
PURPOSE OF REVIEW Metabolism of lipids and lipoproteins, including high-density lipoprotein (HDL), plays a central role in energy homeostasis. Mechanisms underlying the relationship between energy homeostasis and HDL however remain poorly studied. RECENT FINDINGS Available evidence reveals that HDL is implicated in energy homeostasis. Circulating high-density lipoprotein-cholesterol (HDL-C) levels are affected by energy production, raising with increasing resting metabolic rate. Lipolysis of triglycerides as a source of energy decreases plasma levels of remnant cholesterol, increases levels of HDL-C, and can be cardioprotective. Switch to preferential energy production from carbohydrates exerts opposite effects. SUMMARY Low HDL-C may represent a biomarker of inefficient energy production from fats. HDL-C-raising can be beneficial when it reflects enhanced energy production from burning fat.
Collapse
Affiliation(s)
- Anatol Kontush
- Sorbonne University, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), UMR_S 1166, Paris, France
| | - Maximiliano Martin
- Laboratory of Lipids and Atherosclerosis, Department of Clinical Biochemistry, INFIBIOC, University of Buenos Aires. CONICET, Buenos Aires, Argentina
| | - Fernando Brites
- Laboratory of Lipids and Atherosclerosis, Department of Clinical Biochemistry, INFIBIOC, University of Buenos Aires. CONICET, Buenos Aires, Argentina
| |
Collapse
|
26
|
Qu W, Zhou X, Jiang X, Xie X, Xu K, Gu X, Na R, Piao M, Xi X, Sun N, Wang X, Peng X, Xu J, Tian J, Zhang J, Guo J, Zhang M, Zhang Y, Pan Z, Wang K, Yu B, Sun B, Li S, Tian J. Long Noncoding RNA Gpr137b-ps Promotes Advanced Atherosclerosis via the Regulation of Autophagy in Macrophages. Arterioscler Thromb Vasc Biol 2023; 43:e468-e489. [PMID: 37767704 DOI: 10.1161/atvbaha.123.319037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Current therapies cannot completely reverse advanced atherosclerosis. High levels of amino acids, induced by Western diet, stimulate mTORC1 (mammalian target of rapamycin complex 1)-autophagy defects in macrophages, accelerating atherosclerotic plaque progression. In addition, autophagy-lysosomal dysfunction contributes to plaque necrotic core enlargement and lipid accumulation. Therefore, it is essential to investigate the novel mechanism and molecules to reverse amino acid-mTORC1-autophagy signaling dysfunction in macrophages of patients with advanced atherosclerosis. METHODS We observed that Gpr137b-ps (G-protein-coupled receptor 137B, pseudogene) was upregulated in advanced atherosclerotic plaques. The effect of Gpr137b-ps on the progression of atherosclerosis was studied by generating advanced plaques in ApoE-/- mice with cardiac-specific knockout of Gpr137b-ps. Bone marrow-derived macrophages and mouse mononuclear macrophage cell line RAW264.7 cells were subjected to starvation or amino acid stimulation to study amino acid-mTORC1-autophagy signaling. Using both gain- and loss-of-function approaches, we explored the mechanism of Gpr137b-ps-regulated autophagy. RESULTS Our results demonstrated that Gpr137b-ps deficiency led to enhanced autophagy in macrophages and reduced atherosclerotic lesions, characterized by fewer necrotic cores and less lipid accumulation. Knockdown of Gpr137b-ps increased autophagy and prevented amino acid-induced mTORC1 signaling activation. As the downstream binding protein of Gpr137b-ps, HSC70 (heat shock cognate 70) rescued the impaired autophagy induced by Gpr137b-ps. Furthermore, Gpr137b-ps interfered with the HSC70 binding to G3BP (Ras GTPase-activating protein-binding protein), which tethers the TSC (tuberous sclerosis complex) complex to lysosomes and suppresses mTORC1 signaling. In addition to verifying that the NTF2 (nuclear transport factor 2) domain of G3BP binds to HSC70 by in vitro protein synthesis, we further demonstrated that HSC70 binds to the NTF2 domain of G3BP through its W90-F92 motif by using computational modeling. CONCLUSIONS These findings reveal that Gpr137b-ps plays an essential role in the regulation of macrophage autophagy, which is crucial for the progression of advanced atherosclerosis. Gpr137b-ps impairs the interaction of HSC70 with G3BP to regulate amino acid-mTORC1-autophagy signaling, and these results provide a new potential therapeutic direction for the treatment of advanced atherosclerosis.
Collapse
Affiliation(s)
- Wenbo Qu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xin Zhou
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xinjian Jiang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xianwei Xie
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China (X. Xie)
| | - Kaijian Xu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xia Gu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Ruisi Na
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Heilongjiang, China (R.N.)
| | - Minghui Piao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xiangwen Xi
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Na Sun
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xueyu Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Xiang Peng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Junyan Xu
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China (J.X.)
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Cardiovascular Diseases Institute of the First Affiliated Hospital, Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, China (J.X., J.G.)
| | - Jiangtian Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Jian Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology (J.Z.)
| | - Junli Guo
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Cardiovascular Diseases Institute of the First Affiliated Hospital, Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, China (J.X., J.G.)
| | - Maomao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Yao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Zhenwei Pan
- College of Pharmacy (Z.P., B.S.), Harbin Medical University, China
| | - Kun Wang
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (K.W.)
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| | - Bin Sun
- College of Pharmacy (Z.P., B.S.), Harbin Medical University, China
| | - Shuijie Li
- Department of Biopharmaceutical Sciences, College of Pharmacy (S.L.), Harbin Medical University, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases Harbin Medical University, China (S.L.)
- Department of Biopharmaceutical Sciences, College of Pharmacy Harbin Medical University, China (S.L.)
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, China (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian)
- The Key Laboratory of Myocardial Ischemia, Ministry of Education (W.Q., X.Z., X.J., K.X., X.G., M.P., X. Xi, N.S., X.W., X.P., Jiangtian Tian, M.Z., Y.Z., B.Y., Jinwei Tian), Harbin Medical University, China
| |
Collapse
|
27
|
Xia D, Liu Y, Wu P, Wei D. Current Advances of Mitochondrial Dysfunction and Cardiovascular Disease and Promising Therapeutic Strategies. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1485-1500. [PMID: 37481069 DOI: 10.1016/j.ajpath.2023.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/16/2023] [Accepted: 06/29/2023] [Indexed: 07/24/2023]
Abstract
Mitochondria are cellular power stations and essential organelles for maintaining cellular homeostasis. Dysfunctional mitochondria have emerged as a key factor in the occurrence and development of cardiovascular disease. This review focuses on advances in the relationship between mitochondrial dysfunction and cardiovascular diseases such as atherosclerosis, heart failure, myocardial ischemia reperfusion injury, and pulmonary arterial hypertension. The clinical value and challenges of mitochondria-targeted strategies, including mitochondria-targeted antioxidants, mitochondrial quality control modulators, mitochondrial function protectors, mitochondrial biogenesis promoters, and recently developed mitochondrial transplants, are also discussed.
Collapse
Affiliation(s)
- Dexiang Xia
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Yue Liu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Peng Wu
- Hengyang Maternal and Child Health Hospital, Hengyang, China
| | - Dangheng Wei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|
28
|
Hu D, Tan M, Lu D, Kleiboeker B, Liu X, Park H, Kravitz AV, Shoghi KI, Tseng YH, Razani B, Ikeda A, Lodhi IJ. TMEM135 links peroxisomes to the regulation of brown fat mitochondrial fission and energy homeostasis. Nat Commun 2023; 14:6099. [PMID: 37773161 PMCID: PMC10541902 DOI: 10.1038/s41467-023-41849-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 09/20/2023] [Indexed: 10/01/2023] Open
Abstract
Mitochondrial morphology, which is controlled by mitochondrial fission and fusion, is an important regulator of the thermogenic capacity of brown adipocytes. Adipose-specific peroxisome deficiency impairs thermogenesis by inhibiting cold-induced mitochondrial fission due to decreased mitochondrial membrane content of the peroxisome-derived lipids called plasmalogens. Here, we identify TMEM135 as a critical mediator of the peroxisomal regulation of mitochondrial fission and thermogenesis. Adipose-specific TMEM135 knockout in mice blocks mitochondrial fission, impairs thermogenesis, and increases diet-induced obesity and insulin resistance. Conversely, TMEM135 overexpression promotes mitochondrial division, counteracts obesity and insulin resistance, and rescues thermogenesis in peroxisome-deficient mice. Mechanistically, thermogenic stimuli promote association between peroxisomes and mitochondria and plasmalogen-dependent localization of TMEM135 in mitochondria, where it mediates PKA-dependent phosphorylation and mitochondrial retention of the fission factor Drp1. Together, these results reveal a previously unrecognized inter-organelle communication regulating mitochondrial fission and energy homeostasis and identify TMEM135 as a potential target for therapeutic activation of BAT.
Collapse
Affiliation(s)
- Donghua Hu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Min Tan
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Dongliang Lu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian Kleiboeker
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Xuejing Liu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Hongsuk Park
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexxai V Kravitz
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Kooresh I Shoghi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Babak Razani
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
29
|
Wang T, Masedunskas A, Willett WC, Fontana L. Vegetarian and vegan diets: benefits and drawbacks. Eur Heart J 2023; 44:3423-3439. [PMID: 37450568 PMCID: PMC10516628 DOI: 10.1093/eurheartj/ehad436] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023] Open
Abstract
Plant-based diets have become increasingly popular thanks to their purported health benefits and more recently for their positive environmental impact. Prospective studies suggest that consuming vegetarian diets is associated with a reduced risk of developing cardiovascular disease (CVD), diabetes, hypertension, dementia, and cancer. Data from randomized clinical trials have confirmed a protective effect of vegetarian diets for the prevention of diabetes and reductions in weight, blood pressure, glycosylated haemoglobin and low-density lipoprotein cholesterol, but to date, no data are available for cardiovascular event rates and cognitive impairment, and there are very limited data for cancer. Moreover, not all plant-based foods are equally healthy. Unhealthy vegetarian diets poor in specific nutrients (vitamin B12, iron, zinc, and calcium) and/or rich in highly processed and refined foods increase morbidity and mortality. Further mechanistic studies are desirable to understand whether the advantages of healthy, minimally processed vegetarian diets represent an all-or-nothing phenomenon and whether consuming primarily plant-based diets containing small quantities of animal products (e.g. pesco-vegetarian or Mediterranean diets) has beneficial, detrimental, or neutral effects on cardiometabolic health outcomes. Further, mechanistic studies are warranted to enhance our understanding about healthy plant-based food patterns and the biological mechanisms linking dietary factors, CVD, and other metabolic diseases.
Collapse
Affiliation(s)
- Tian Wang
- Charles Perkins Center, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Andrius Masedunskas
- Charles Perkins Center, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Walter C Willett
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Luigi Fontana
- Charles Perkins Center, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Department of Clinical and Experimental Sciences, Brescia University, Brescia, Lombardy, Italy
| |
Collapse
|
30
|
Guan L, Liu R. The Role of Diet and Gut Microbiota Interactions in Metabolic Homeostasis. Adv Biol (Weinh) 2023; 7:e2300100. [PMID: 37142556 DOI: 10.1002/adbi.202300100] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/10/2023] [Indexed: 05/06/2023]
Abstract
Diet is a pivotal determinant in shaping the structure and function of resident microorganisms in the gut through different food components, nutritive proportion, and calories. The effects of diet on host metabolism and physiology can be mediated through the gut microbiota. Gut microbiota-derived metabolites have been shown to regulate glucose and lipid metabolism, energy consumption, and the immune system. On the other hand, emerging evidence indicates that baseline gut microbiota could predict the efficacy of diet intervention, highlighting gut microbiota can be harnessed as a biomarker in personalized nutrition. In this review, the alterations of gut microbiota in different dietary components and dietary patterns, and the potential mechanisms in the diet-microbiota crosstalk are summarized to understand the interactions of diet and gut microbiota on the impact of metabolic homeostasis.
Collapse
Affiliation(s)
- Lizhi Guan
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Disease, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the P. R. China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Disease, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the P. R. China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
31
|
Jeevarathinam S, Al Sabei S, Al Wardi Y. Acute Myocardial Infarction in a Young Bodybuilder Fighter Pilot. Aerosp Med Hum Perform 2023; 94:719-722. [PMID: 37587624 DOI: 10.3357/amhp.6271.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
BACKGROUND: Although advanced coronary artery disease in young, healthy fighter pilots is uncommon, an acute cardiac event in flight could be catastrophic.CASE REPORT: After a gym workout, a 31-yr-old F-16 pilot reported severe central chest pain, one vomiting episode, and excessive sweating but no radiation of pain. Electrocardiograph showed ST elevation in V2-V6. Coronary arteriography showed a thrombotic lesion at the proximal left anterior descending (LAD) artery (90%) and one occluded LAD branch with thrombus; the rest of the arteries were normal and ejection fraction was 55%. Primary percutaneous coronary intervention to LAD with one drug-eluting stent was done. The pilot was discharged in stable hemodynamic condition with medication advice. Assessment revealed no significant cardiac risk factors. He did not seek medical care for two central chest pain episodes following a gym workout prior to this event because rest relieved the pain. He gave a history of using commercial protein supplements for bodybuilding in the past 6 yr.DISCUSSION: In this case report, the impact of aggressive gym workouts and chronic use of commercially available bodybuilding protein supplements on cardiovascular health is discussed, as well as aeromedical dilemmas related to this pilot's career. This case sparks debate about whether a highly motivated young pilot with an unexpected cardiac event should be subjected to regular intensive cardiac evaluation throughout his remaining flying career, with permanent flying limitations, or be motivated to pursue a career shift to facilitate noncomplicated career rehabilitation.Jeevarathinam S, Sabei SA, Wardi YA. Acute myocardial infarction in a young bodybuilder fighter pilot. Aerosp Med Hum Perform. 2023; 94(9):719-722.
Collapse
|
32
|
Li X, Xiao Z, Li C, Chen Q, Jia L. Maternal dietary patterns during pregnancy and the risk of infantile eczema during the first year of life: a cohort study in northeast China. BMC Public Health 2023; 23:1641. [PMID: 37641073 PMCID: PMC10463679 DOI: 10.1186/s12889-023-16577-9] [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: 02/24/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND There are few studies on the relationship between diet during pregnancy and infantile eczema and the conclusions are inconsistent. The aim of the present study was to explore the impact of dietary patterns during pregnancy on infantile eczema. METHODS A total of 495 mother-child pairs from a prospective cohort in Shenyang, China was recruited. Information on maternal dietary intake during pregnancy was assessed with a validated self-administered food frequency questionnaire. The data of infantile eczema was assessed using a structured questionnaire. Factor analysis to derive dietary patterns. The relationship between the dietary pattern and infantile eczema was examined by the logistic regression analysis. RESULTS The cumulative incidence of eczema in 6 months and 12 months in northeast China was 45.7% and 57.8%, respectively. Three dietary patterns were identified. There was a tendency for an expose-response relationship between the maternal high-protein dietary pattern during pregnancy and the risk of infantile eczema within 12 months (P for trend = 0.023): the adjusted odds ratio (95% confidence interval) in the Q1, Q2, Q3, Q4 were 1.00 (reference), 1.63 (0.96-2.76), 1.81 (1.06-3.06), and 1.87 (1.09-3.20), respectively. No association between Western and plant-based patterns during pregnancy and infantile eczema within 12 months was found. Infantile eczema within 6 months was not associated with any of the three dietary patterns. CONCLUSION The maternal high-protein pattern during pregnancy may be a risk factor for infantile eczema during the first year of life.
Collapse
Affiliation(s)
- Xuening Li
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
- Department of Pediatrics, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning, China
| | - Zhe Xiao
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Chenyang Li
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Qi Chen
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Lihong Jia
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China.
- Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Shenyang 110122, Liaoning, China.
| |
Collapse
|
33
|
Zhang X, Evans TD, Chen S, Sergin I, Stitham J, Jeong SJ, Rodriguez-Velez A, Yeh YS, Park A, Jung IH, Diwan A, Schilling JD, Rom O, Yurdagul A, Epelman S, Cho J, Lodhi IJ, Mittendorfer B, Razani B. Loss of Macrophage mTORC2 Drives Atherosclerosis via FoxO1 and IL-1β Signaling. Circ Res 2023; 133:200-219. [PMID: 37350264 PMCID: PMC10527041 DOI: 10.1161/circresaha.122.321542] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 06/12/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND The mTOR (mechanistic target of rapamycin) pathway is a complex signaling cascade that regulates cellular growth, proliferation, metabolism, and survival. Although activation of mTOR signaling has been linked to atherosclerosis, its direct role in lesion progression and in plaque macrophages remains poorly understood. We previously demonstrated that mTORC1 (mTOR complex 1) activation promotes atherogenesis through inhibition of autophagy and increased apoptosis in macrophages. METHODS Using macrophage-specific Rictor- and mTOR-deficient mice, we now dissect the distinct functions of mTORC2 pathways in atherogenesis. RESULTS In contrast to the atheroprotective effect seen with blockade of macrophage mTORC1, macrophage-specific mTORC2-deficient mice exhibit an atherogenic phenotype, with larger, more complex lesions and increased cell death. In cultured macrophages, we show that mTORC2 signaling inhibits the FoxO1 (forkhead box protein O1) transcription factor, leading to suppression of proinflammatory pathways, especially the inflammasome/IL (interleukin)-1β response, a key mediator of vascular inflammation and atherosclerosis. In addition, administration of FoxO1 inhibitors efficiently rescued the proinflammatory response caused by mTORC2 deficiency both in vitro and in vivo. Interestingly, collective deletion of macrophage mTOR, which ablates mTORC1- and mTORC2-dependent pathways, leads to minimal change in plaque size or complexity, reflecting the balanced yet opposing roles of these signaling arms. CONCLUSIONS Our data provide the first mechanistic details of macrophage mTOR signaling in atherosclerosis and suggest that therapeutic measures aimed at modulating mTOR need to account for its dichotomous functions.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Trent D. Evans
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Sunny Chen
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Ismail Sergin
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Jeremiah Stitham
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | - Se-Jin Jeong
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | | | - Yu-Sheng Yeh
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Arick Park
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - In-Hyuk Jung
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Abhinav Diwan
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St. Louis, MO, USA
| | - Joel D. Schilling
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Oren Rom
- Department of Pathology and Translational Pathobiology and Department of Molecular and Cellular Physiology, Louisiana State University, Shreveport, LA
| | - Arif Yurdagul
- Department of Pathology and Translational Pathobiology and Department of Molecular and Cellular Physiology, Louisiana State University, Shreveport, LA
| | - Slava Epelman
- Ted Rogers Centre for Heart Research, Peter Munk Cardiac Center, Toronto General Hospital Research Institute, University Health Network and University of Toronto, Toronto, Canada
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Irfan J. Lodhi
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | - Bettina Mittendorfer
- Division of Geriatrics and Nutritional Science, and Washington University School of Medicine, St Louis, MO, USA
| | - Babak Razani
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Pittsburgh VA Medical Center, Pittsburgh, PA
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St. Louis, MO, USA
| |
Collapse
|
34
|
Shoja M, Borazjani F, Ahmadi Angali K, Hosseini SA, Hashemi SJ. The dietary patterns derived by reduced-rank regression in association with Framingham risk score and lower DASH score in Hoveyzeh cohort study. Sci Rep 2023; 13:11093. [PMID: 37422506 PMCID: PMC10329634 DOI: 10.1038/s41598-023-37809-3] [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: 10/11/2022] [Accepted: 06/28/2023] [Indexed: 07/10/2023] Open
Abstract
The relationship between dietary patterns (DPs) and cardiovascular disease (CVD) has been the subject of much research, but given the significance of this disease, studying the factors affecting it through different methodological considerations is of utmost importance. This study aimed to investigate the association between the four dietary patterns (DPs) derived from reduced-rank regression (RRR) and the risk of CVD predicted by the Framingham Risk Score (FRS) in the Arab residence of Khuzestan, Iran. Furthermore, the predefined Dietary Approaches to Stop Hypertension (DASH) would be used as a comparative model to assess the validity of the extracted DPs. In this cross-sectional study, 5799 individuals aged 35-70 without a CVD diagnosis were selected among the participants of the Hoveyzeh cohort study (HCS). The Risk of CVD was assessed using the FRS model. A semi-quantitative food frequency questionnaire evaluated dietary intake. Four DPs were derived using RRR with 28 food groups as predictors and total protein (g/d), fiber(g/d), fat(g/d), and magnesium intake (mg/d) as response variables. Multinomial and binary logistic regression were used to assess the relationship of DPs with intermediate (10-20%) and high (> 20%) levels of FRS and lower DASH scores (< 4.5), respectively. Four primary DPs were derived, which explained 89.10 of the total explained variance in participants' dietary intake. Multinomial regression was applied between FRS (10-20%) and (> 20%) across quartiles of four identified DPs. After adjustment for potential confounders, higher tendency to 1st and 2nd DPs in Model 1, OR = 4.67 (95% CI 3.65; 6.01), OR = 1.42 (95% CI 1.13; 1.79) were presented accordingly. The 1st DP, characterized by higher intake of refined grains and lower intake of vegetables oil, sugar, mayonnaise and artificial juices, the 2nd DP characterized by higher intake of hydrogenated fat and lower consumption of tomato sauce and soft drink was associated with greater odds of CVD with the intermediate level of FRS. However, higher adherence to the 3rd DP, characterized by higher intake of fruits, vegetables and legumes and lower intake of fish, egg, red meat, processed meat, mayonnaise, sugar and artificial juices, the 4th DP characterized by higher intake of coffee, nuts and lower intake of sugar, mayonnaise and artificial juices was associated with a lower risk of FRS. Moreover, lower DASH score considered in binary logistic regression across quartiles of four identified dietary patterns. 1st and 2nd DPs were directly related to lower DASH scores, while 3rd and 4th DPs had high comparability with the DASH diet and inversely contributed to the lower DASH score. Total DASH score was significantly correlated to four derived DPs. Our findings confirm the current knowledge regarding the beneficial effects of healthy plant-based DPs and the avoidance of high-fat and processed foods to prevent CVD.
Collapse
Affiliation(s)
- Marzieh Shoja
- Nutrition and Metabolic Diseases Research Center and clinical sciences research institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Borazjani
- Nutrition and Metabolic Diseases Research Center and clinical sciences research institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Nutrition, Faculty of Allied Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Kambiz Ahmadi Angali
- Department of Biostatistics, School of Health Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Ahmad Hosseini
- Nutrition and Metabolic Diseases Research Center and clinical sciences research institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Jalal Hashemi
- Alimentary Tract Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
35
|
Desai JV, Kumar D, Freiwald T, Chauss D, Johnson MD, Abers MS, Steinbrink JM, Perfect JR, Alexander B, Matzaraki V, Snarr BD, Zarakas MA, Oikonomou V, Silva LM, Shivarathri R, Beltran E, Demontel LN, Wang L, Lim JK, Launder D, Conti HR, Swamydas M, McClain MT, Moutsopoulos NM, Kazemian M, Netea MG, Kumar V, Köhl J, Kemper C, Afzali B, Lionakis MS. C5a-licensed phagocytes drive sterilizing immunity during systemic fungal infection. Cell 2023; 186:2802-2822.e22. [PMID: 37220746 PMCID: PMC10330337 DOI: 10.1016/j.cell.2023.04.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 03/10/2023] [Accepted: 04/21/2023] [Indexed: 05/25/2023]
Abstract
Systemic candidiasis is a common, high-mortality, nosocomial fungal infection. Unexpectedly, it has emerged as a complication of anti-complement C5-targeted monoclonal antibody treatment, indicating a critical niche for C5 in antifungal immunity. We identified transcription of complement system genes as the top biological pathway induced in candidemic patients and as predictive of candidemia. Mechanistically, C5a-C5aR1 promoted fungal clearance and host survival in a mouse model of systemic candidiasis by stimulating phagocyte effector function and ERK- and AKT-dependent survival in infected tissues. C5ar1 ablation rewired macrophage metabolism downstream of mTOR, promoting their apoptosis and enhancing mortality through kidney injury. Besides hepatocyte-derived C5, local C5 produced intrinsically by phagocytes provided a key substrate for antifungal protection. Lower serum C5a concentrations or a C5 polymorphism that decreases leukocyte C5 expression correlated independently with poor patient outcomes. Thus, local, phagocyte-derived C5 production licenses phagocyte antimicrobial function and confers innate protection during systemic fungal infection.
Collapse
Affiliation(s)
- Jigar V Desai
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA
| | - Dhaneshwar Kumar
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA; Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Tilo Freiwald
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Daniel Chauss
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | | | - Michael S Abers
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA
| | - Julie M Steinbrink
- Department of Medicine, Division of Infectious Diseases, Duke University, Durham, NC, USA
| | - John R Perfect
- Department of Medicine, Division of Infectious Diseases, Duke University, Durham, NC, USA
| | - Barbara Alexander
- Department of Medicine, Division of Infectious Diseases, Duke University, Durham, NC, USA
| | - Vasiliki Matzaraki
- Department of Genetics, University of Groningen, Groningen, the Netherlands
| | - Brendan D Snarr
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA
| | - Marissa A Zarakas
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA
| | - Vasileios Oikonomou
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA
| | - Lakmali M Silva
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD, USA
| | - Raju Shivarathri
- Center for Discovery & Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Emily Beltran
- Complement and Inflammation Research Section, National Heart Lung and Blood Institute, NIH, Bethesda, MD, USA
| | - Luciana Negro Demontel
- Complement and Inflammation Research Section, National Heart Lung and Blood Institute, NIH, Bethesda, MD, USA
| | - Luopin Wang
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Jean K Lim
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dylan Launder
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Heather R Conti
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Muthulekha Swamydas
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA
| | - Micah T McClain
- Department of Medicine, Division of Infectious Diseases, Duke University, Durham, NC, USA
| | - Niki M Moutsopoulos
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD, USA
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University, Nijmegen, the Netherlands
| | - Vinod Kumar
- Department of Genetics, University of Groningen, Groningen, the Netherlands; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University, Nijmegen, the Netherlands
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Claudia Kemper
- Complement and Inflammation Research Section, National Heart Lung and Blood Institute, NIH, Bethesda, MD, USA
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy & Infectious Diseases, NIH, Bethesda, MD, USA.
| |
Collapse
|
36
|
Shang Y, Li Z, Cai P, Li W, Xu Y, Zhao Y, Xia S, Shao Q, Wang H. Megamitochondria plasticity: function transition from adaption to disease. Mitochondrion 2023:S1567-7249(23)00053-3. [PMID: 37276954 DOI: 10.1016/j.mito.2023.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/08/2023] [Accepted: 06/02/2023] [Indexed: 06/07/2023]
Abstract
As the cell's energy factory and metabolic hub, mitochondria are critical for ATP synthesis to maintain cellular function. Mitochondria are highly dynamic organelles that continuously undergo fusion and fission to alter their size, shape, and position, with mitochondrial fusion and fission being interdependent to maintain the balance of mitochondrial morphological changes. However, in response to metabolic and functional damage, mitochondria can grow in size, resulting in a form of abnormal mitochondrial morphology known as megamitochondria. Megamitochondria are characterized by their considerably larger size, pale matrix, and marginal cristae structure and have been observed in various human diseases. In energy-intensive cells like hepatocytes or cardiomyocytes, the pathological process can lead to the growth of megamitochondria, which can further cause metabolic disorders, cell damage and aggravates the progression of the disease. Nonetheless, megamitochondria can also form in response to short-term environmental stimulation as a compensatory mechanism to support cell survival. However, extended stimulation can reverse the benefits of megamitochondria leading to adverse effects. In this review, we will focus on the findings of the different roles of megamitochondria, and their link to disease development to identify promising clinical therapeutic targets.
Collapse
Affiliation(s)
- Yuxing Shang
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Zhanghui Li
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Peiyang Cai
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Wuhao Li
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Ye Xu
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Yangjing Zhao
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Sheng Xia
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Qixiang Shao
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China; Institute of Medical Genetics and Reproductive Immunity, School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai'an 223002, Jiangsu, PR China.
| | - Hui Wang
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China.
| |
Collapse
|
37
|
Klatt KC, Bass K, Speakman JR, Hall KD. Chowing down: diet considerations in rodent models of metabolic disease. LIFE METABOLISM 2023; 2:load013. [PMID: 37485302 PMCID: PMC10361708 DOI: 10.1093/lifemeta/load013] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Diet plays a substantial role in the etiology, progression, and treatment of chronic disease and is best considered as a multifaceted set of modifiable input variables with pleiotropic effects on a variety of biological pathways spanning multiple organ systems. This brief review discusses key issues related to the design and conduct of diet interventions in rodent models of metabolic disease and their implications for interpreting experiments. We also make specific recommendations to improve rodent diet studies to help better understand the role of diet on metabolic physiology and thereby improve our understanding of metabolic disease.
Collapse
Affiliation(s)
- Kevin C. Klatt
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Kevin Bass
- Garrison Institute of Aging, Texas Tech University Health Science Center, Lubbock, TX 79430, USA
| | - John R. Speakman
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Kevin D. Hall
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
38
|
Zhang Y, Weng J, Huan L, Sheng S, Xu F. Mitophagy in atherosclerosis: from mechanism to therapy. Front Immunol 2023; 14:1165507. [PMID: 37261351 PMCID: PMC10228545 DOI: 10.3389/fimmu.2023.1165507] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/12/2023] [Indexed: 06/02/2023] Open
Abstract
Mitophagy is a type of autophagy that can selectively eliminate damaged and depolarized mitochondria to maintain mitochondrial activity and cellular homeostasis. Several pathways have been found to participate in different steps of mitophagy. Mitophagy plays a significant role in the homeostasis and physiological function of vascular endothelial cells, vascular smooth muscle cells, and macrophages, and is involved in the development of atherosclerosis (AS). At present, many medications and natural chemicals have been shown to alter mitophagy and slow the progression of AS. This review serves as an introduction to the field of mitophagy for researchers interested in targeting this pathway as part of a potential AS management strategy.
Collapse
Affiliation(s)
- Yanhong Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiajun Weng
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University, Beijing, China
- Department of Integrated Traditional and Western Medicine, Peking University Health Science Center, Beijing, China
| | - Luyao Huan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Song Sheng
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengqin Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University, Beijing, China
- Department of Integrated Traditional and Western Medicine, Peking University Health Science Center, Beijing, China
| |
Collapse
|
39
|
Barbero NM, Oller J, Sanz AB, Ramos AM, Ortiz A, Ruiz-Ortega M, Rayego-Mateos S. Mitochondrial Dysfunction in the Cardio-Renal Axis. Int J Mol Sci 2023; 24:ijms24098209. [PMID: 37175915 PMCID: PMC10179675 DOI: 10.3390/ijms24098209] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Cardiovascular disease (CVD) frequently complicates chronic kidney disease (CKD). The risk of all-cause mortality increases from 20% to 500% in patients who suffer both conditions; this is referred to as the so-called cardio-renal syndrome (CRS). Preclinical studies have described the key role of mitochondrial dysfunction in cardiovascular and renal diseases, suggesting that maintaining mitochondrial homeostasis is a promising therapeutic strategy for CRS. In this review, we explore the malfunction of mitochondrial homeostasis (mitochondrial biogenesis, dynamics, oxidative stress, and mitophagy) and how it contributes to the development and progression of the main vascular pathologies that could be affected by kidney injury and vice versa, and how this knowledge may guide the development of novel therapeutic strategies in CRS.
Collapse
Affiliation(s)
- Nerea Mendez Barbero
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, 28037 Madrid, Spain
| | - Jorge Oller
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, 28037 Madrid, Spain
| | - Ana B Sanz
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Adrian M Ramos
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Alberto Ortiz
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Marta Ruiz-Ortega
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
| | - Sandra Rayego-Mateos
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
| |
Collapse
|
40
|
Fu W, Wu G. Targeting mTOR for Anti-Aging and Anti-Cancer Therapy. Molecules 2023; 28:molecules28073157. [PMID: 37049920 PMCID: PMC10095787 DOI: 10.3390/molecules28073157] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
The balance between anabolism and catabolism is disrupted with aging, with the rate of anabolism being faster than that of catabolism. Therefore, mTOR, whose major function is to enhance anabolism and inhibit catabolism, has become a potential target of inhibition for anti-aging therapy. Interestingly, it was found that the downregulation of the mTOR signaling pathway had a lifespan-extending effect resembling calorie restriction. In addition, the mTOR signaling pathway promotes cell proliferation and has been regarded as a potential anti-cancer target. Rapamycin and rapalogs, such as everolimus, have proven to be effective in preventing certain tumor growth. Here, we reviewed the basic knowledge of mTOR signaling, including both mTORC1 and mTORC2. Then, for anti-aging, we cited a lot of evidence to discuss the role of targeting mTOR and its anti-aging mechanism. For cancer therapy, we also discussed the role of mTOR signaling in different types of cancers, including idiopathic pulmonary fibrosis, tumor immunity, etc. In short, we discussed the research progress and both the advantages and disadvantages of targeting mTOR in anti-aging and anti-cancer therapy. Hopefully, this review may promote more ideas to be generated for developing inhibitors of mTOR signaling to fight cancer and extend lifespan.
Collapse
Affiliation(s)
- Wencheng Fu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, the Joint International Research Laboratory of Metabolic & Developmental Sciences MOE, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Geng Wu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, the Joint International Research Laboratory of Metabolic & Developmental Sciences MOE, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
41
|
Jing W, Chen C, Wang G, Han M, Chen S, Jiang X, Shi C, Sun P, Yang Z, Shi B, Jiang X. Metabolic Modulation of Intracellular Ammonia via Intravesical Instillation of Nanoporter-Encased Hydrogel Eradicates Bladder Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206893. [PMID: 36775865 PMCID: PMC10131795 DOI: 10.1002/advs.202206893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/25/2023] [Indexed: 06/18/2023]
Abstract
Tumor protein 53 (TP53) mutation in bladder carcinoma (BC), upregulates the transcription of carbamoyl phosphate synthetase 1 (CPS1), to reduce intracellular ammonia toxicity. To leverage ammonia combating BC, here, an intravesically perfusable nanoporter-encased hydrogel system is reported. A biomimetic fusogenic liposomalized nanoporter (FLNP) that is decorated with urea transporter-B (UT-B) is first synthesized with protonated chitosan oligosaccharide for bladder tumor-targeted co-delivery of urease and small interfering RNA targeting CPS1 (siCPS1). Mussel-inspired hydrogel featured with dual functions of bio-adhesion and injectability is then fabricated as the reservoir for intravesical immobilization of FLNP. It is found that FLNP-mediated UT-B immobilization dramatically induces urea transportation into tumor cells, and co-delivery of urease and siCPS1 significantly boosts ammonia accumulation in tumor inducing cell apoptosis. Treatment with hybrid system exhibits superior anti-tumor effect in orthotopic bladder tumor mouse model and patient-derived xenograft model, respectively. Combined with high-protein diet, the production of urinary urea increases, leading to an augmented intracellular deposition of ammonia in BC cells, and ultimately an enhanced tumor inhibition. Together, the work establishes that cascade modulation of ammonia in tumor cells could induce tumor apoptosis and may be a practical strategy for eradication of TP53-mutated bladder cancer.
Collapse
Affiliation(s)
- Weiqiang Jing
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Chen Chen
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Ganyu Wang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Maosen Han
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Shouzhen Chen
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Xin Jiang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Chongdeng Shi
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Peng Sun
- Shandong University of Traditional Chinese Medicine, University Road, Jinan, Shandong Province, 250355, China
| | - Zhenmei Yang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| | - Xinyi Jiang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Cultural West Road, Jinan, Shandong Province, 250012, China
| |
Collapse
|
42
|
Mantzouranis E, Kakargia E, Kakargias F, Lazaros G, Tsioufis K. The Impact of High Protein Diets on Cardiovascular Outcomes: A Systematic Review and Meta-Analysis of Prospective Cohort Studies. Nutrients 2023; 15:1372. [PMID: 36986102 PMCID: PMC10058321 DOI: 10.3390/nu15061372] [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: 02/16/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/14/2023] Open
Abstract
High protein diets have gained increased popularity as a means of losing weight, increasing muscle mass and strength, and improving cardiometabolic parameters. Only a few meta-analyses have addressed their impact on cardiovascular morbidity and mortality and failed to show any significant associations without applying strict values to define high protein intake. Due to the conflicting research background, we conducted a meta-analysis to assess the impact of high protein diets compared to normal protein consumption on cardiovascular outcomes in adults without established cardiovascular disease. Fourteen prospective cohort studies were included. A total of 6 studies, including 221,583 participants, reported data about cardiovascular death, without showing a statistically significant difference in the random effect model (odds ratio: 0.94; confidence interval: 0.60-1.46; I2 = 98%; p = 0.77). Analysis of three studies, which included 90,231 participants showed that a high protein diet was not associated with a lower risk of stroke (odds ratio: 1.02; confidence interval: 0.94-1.10; I2 = 0%; p = 0.66). Regarding the secondary outcome of non-fatal myocardial infarction, stroke, or cardiovascular death, 13 studies that included 525,047 participants showed no statistically significant difference (odds ratio; 0.87; confidence interval: 0.70-1.07; I2 = 97%; p = 0.19). In conclusion, according to our study results, high protein consumption does not affect cardiovascular prognosis.
Collapse
Affiliation(s)
- Emmanouil Mantzouranis
- 1st Cardiology Clinic, Hippokration Hospital, University of Athens, 115 27 Athens, Greece
| | - Eleftheria Kakargia
- Internal Medicine Clinic, 401 General Military Hospital, 115 25 Athens, Greece
| | - Fotis Kakargias
- Internal Medicine Clinic, 401 General Military Hospital, 115 25 Athens, Greece
| | - George Lazaros
- 1st Cardiology Clinic, Hippokration Hospital, University of Athens, 115 27 Athens, Greece
| | - Konstantinos Tsioufis
- 1st Cardiology Clinic, Hippokration Hospital, University of Athens, 115 27 Athens, Greece
| |
Collapse
|
43
|
Zhang X, Misra SK, Moitra P, Zhang X, Jeong SJ, Stitham J, Rodriguez-Velez A, Park A, Yeh YS, Gillanders WE, Fan D, Diwan A, Cho J, Epelman S, Lodhi IJ, Pan D, Razani B. Use of acidic nanoparticles to rescue macrophage lysosomal dysfunction in atherosclerosis. Autophagy 2023; 19:886-903. [PMID: 35982578 PMCID: PMC9980706 DOI: 10.1080/15548627.2022.2108252] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/19/2022] Open
Abstract
Dysfunction in the macrophage lysosomal system including reduced acidity and diminished degradative capacity is a hallmark of atherosclerosis, leading to blunted clearance of excess cellular debris and lipids in plaques and contributing to lesion progression. Devising strategies to rescue this macrophage lysosomal dysfunction is a novel therapeutic measure. Nanoparticles have emerged as an effective platform to both target specific tissues and serve as drug delivery vehicles. In most cases, administered nanoparticles are taken up non-selectively by the mononuclear phagocyte system including monocytes/macrophages leading to the undesirable degradation of cargo in lysosomes. We took advantage of this default route to target macrophage lysosomes to rectify their acidity in disease states such as atherosclerosis. Herein, we develop and test two commonly used acidic nanoparticles, poly-lactide-co-glycolic acid (PLGA) and polylactic acid (PLA), both in vitro and in vivo. Our results in cultured macrophages indicate that the PLGA-based nanoparticles are the most effective at trafficking to and enhancing acidification of lysosomes. PLGA nanoparticles also provide functional benefits including enhanced lysosomal degradation, promotion of macroautophagy/autophagy and protein aggregate removal, and reduced apoptosis and inflammasome activation. We demonstrate the utility of this system in vivo, showing nanoparticle accumulation in, and lysosomal acidification of, macrophages in atherosclerotic plaques. Long-term administration of PLGA nanoparticles results in significant reductions in surrogates of plaque complexity with reduced apoptosis, necrotic core formation, and cytotoxic protein aggregates and increased fibrous cap formation. Taken together, our data support the use of acidic nanoparticles to rescue macrophage lysosomal dysfunction in the treatment of atherosclerosis.Abbreviations: BCA: brachiocephalic arteries; FACS: fluorescence activated cell sorting; FITC: fluorescein-5-isothiocyanatel; IL1B: interleukin 1 beta; LAMP: lysosomal associated membrane protein; LIPA/LAL: lipase A, lysosomal acid type; LSDs: lysosomal storage disorders; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MFI: mean fluorescence intensity; MPS: mononuclear phagocyte system; PEGHDE: polyethylene glycol hexadecyl ether; PLA: polylactic acid; PLGA: poly-lactide-co-glycolic acid; SQSTM1/p62: sequestosome 1.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Cardiovascular Division, Washington University, St. Louis, MO, USA
| | - Santosh Kumar Misra
- Department of Bioengineering, University of Illinois at Urbana Champaign, IL, USA
| | - Parikshit Moitra
- Departments of Diagnostic Radiology and Nuclear Medicine and Pediatrics, Baltimore, Maryland, USA
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania16802, USA
| | - Xiuli Zhang
- Department of Surgery, Washington University, St. Louis, MO, USA
| | - Se-Jin Jeong
- Cardiovascular Division, Washington University, St. Louis, MO, USA
| | - Jeremiah Stitham
- Cardiovascular Division, Washington University, St. Louis, MO, USA
- Division of Endocrinology, Metabolism, and Lipid Research, St. Louis, MO, USA
| | | | - Arick Park
- Cardiovascular Division, Washington University, St. Louis, MO, USA
| | - Yu-Sheng Yeh
- Cardiovascular Division, Washington University, St. Louis, MO, USA
| | | | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Abhinav Diwan
- Cardiovascular Division, Washington University, St. Louis, MO, USA
- John Cochran Division, VA Medical Center, St. Louis, MO, USA
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Slava Epelman
- Peter Munk Cardiac Center, Toronto General Hospital Research Institute, University Health Network, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Irfan J. Lodhi
- Division of Endocrinology, Metabolism, and Lipid Research, St. Louis, MO, USA
| | - Dipanjan Pan
- Department of Bioengineering, University of Illinois at Urbana Champaign, IL, USA
- Departments of Diagnostic Radiology and Nuclear Medicine and Pediatrics, Baltimore, Maryland, USA
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania16802, USA
| | - Babak Razani
- Cardiovascular Division, Washington University, St. Louis, MO, USA
- John Cochran Division, VA Medical Center, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
44
|
Xu D, Xu Y, Zhang B, Wang Y, Han L, Sun J, Sun H. Higher dietary intake of aromatic amino acids was associated with lower risk of cardiovascular disease mortality in adult participants in NHANES III. Nutr Res 2023; 113:39-48. [PMID: 37023498 DOI: 10.1016/j.nutres.2023.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/08/2023] [Accepted: 03/07/2023] [Indexed: 03/19/2023]
Abstract
Little is known about the associations between dietary aromatic amino acids (AAAs) intake and mortality from all causes and cardiovascular disease (CVD). Accordingly, we evaluated these associations in the adult population of the United States using data from the Third National Health and Nutrition Examination Survey. This was a cohort study. Dietary intake of AAAs (tyrosine, phenylalanine, and tryptophan) was determined from the total nutrient intake document. We hypothesized that higher dietary AAA intake would lower all-cause and CVD mortality in adults in the United States. First, we categorized participants into quintiles based on their dietary intakes of total AAAs, tyrosine, phenylalanine, and tryptophan. Then, we established 4 Cox proportional-hazards models (models 1-4) and calculated hazard ratios and 95% confidence intervals to estimate the associations between dietary intakes of total AAAs, tyrosine, phenylalanine, and tryptophan and all-cause and CVD mortality. Mortality status was primarily obtained from files linked to the National Death Index records up to December 31, 2015. After multivariate adjustment, the hazard ratios (95% confidence intervals) of CVD mortality in the highest quintiles of dietary total AAAs, tyrosine, phenylalanine, and tryptophan intake (reference: the lowest quintiles) were 0.66 (0.52-0.84), 0.65 (0.51-0.83), 0.66 (0.52-0.85) and 0.64 (0.50-0.82), respectively. In a nationally representative cohort, higher dietary intakes of total AAA and the 3 individual AAAs were independently associated with a lower risk of CVD mortality, and these associations were stronger in non-Hispanic White people than in other people.
Collapse
|
45
|
Wu Q, Lv Q, Liu X, Ye X, Cao L, Wang M, Li J, Yang Y, Li L, Wang S. Natural compounds from botanical drugs targeting mTOR signaling pathway as promising therapeutics for atherosclerosis: A review. Front Pharmacol 2023; 14:1083875. [PMID: 36744254 PMCID: PMC9894899 DOI: 10.3389/fphar.2023.1083875] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/05/2023] [Indexed: 01/22/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease that is a major cause of cardiovascular diseases (CVDs), including coronary artery disease, hypertension, myocardial infarction, and heart failure. Hence, the mechanisms of AS are still being explored. A growing compendium of evidence supports that the activity of the mechanistic/mammalian target of rapamycin (mTOR) is highly correlated with the risk of AS. The mTOR signaling pathway contributes to AS progression by regulating autophagy, cell senescence, immune response, and lipid metabolism. Various botanical drugs and their functional compounds have been found to exert anti- AS effects by modulating the activity of the mTOR signaling pathway. In this review, we summarize the pathogenesis of AS based on the mTOR signaling pathway from the aspects of immune response, autophagy, cell senescence, and lipid metabolism, and comb the recent advances in natural compounds from botanical drugs to inhibit the mTOR signaling pathway and delay AS development. This review will provide a new perspective on the mechanisms and precision treatments of AS.
Collapse
Affiliation(s)
- Qian Wu
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Qianyu Lv
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao’an Liu
- Capital University of Medical, Beijing, China
| | - Xuejiao Ye
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Linlin Cao
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Manshi Wang
- Beijing Xicheng District Guangwai Hospital, Beijing, China
| | - Junjia Li
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Yingtian Yang
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Lanlan Li
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Shihan Wang
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
46
|
Liu C, Li Z, Li B, Liu W, Zhang S, Qiu K, Zhu W. Relationship between ferroptosis and mitophagy in cardiac ischemia reperfusion injury: a mini-review. PeerJ 2023; 11:e14952. [PMID: 36935924 PMCID: PMC10019339 DOI: 10.7717/peerj.14952] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 02/03/2023] [Indexed: 03/14/2023] Open
Abstract
Cardiovascular diseases (CVD), with high morbidity and mortality, seriously affect people's life and social development. Clinically, reperfusion therapy is typically used to treat ischemic cardiomyopathy, such as severe coronary heart disease and acute myocardial infarction. However, reperfusion therapy can lead to myocardial ischemia reperfusion injury (MIRI), which can affect the prognosis of patients. Studying the mechanisms of MIRI can help us improve the treatment of MIRI. The pathological process of MIRI involves many mechanisms such as ferroptosis and mitophagy. Ferroptosis can exacerbate MIRI, and regulation of mitophagy can alleviate MIRI. Both ferroptosis and mitophagy are closely related to ROS, but there is no clear understanding of the relationship between ferroptosis and mitophagy. In this review, we analyzed the relationship between ferroptosis and mitophagy according to the role of mTOR, NLPR3 and HIF. In addition, simultaneous regulation of mitophagy and ferroptosis may be superior to single therapy for MIRI. We summarized potential drugs that can regulate mitophagy and/or ferroptosis, hoping to provide reference for the development of drugs and methods for MIRI treatment.
Collapse
Affiliation(s)
- Cuihua Liu
- Third-Grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, Hubei Province, China
| | - Zunjiang Li
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Botao Li
- Third-Grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, Hubei Province, China
| | - Wei Liu
- Third-Grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, Hubei Province, China
| | - Shizhong Zhang
- Third-Grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, Hubei Province, China
| | - Kuncheng Qiu
- Third-Grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang, Hubei Province, China
| | - Wei Zhu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| |
Collapse
|
47
|
Zhang X, Stitham J, Rodriguez-Velez A, Jeong SJ, Park A, Yeh YS, Kapoor D, Mittendorfer B, Razani B. Evaluation of mTORC1 signaling in mouse atherosclerotic macrophages by flow cytometry and immunofluorescence. STAR Protoc 2022; 3:101665. [PMID: 36094885 PMCID: PMC9474552 DOI: 10.1016/j.xpro.2022.101665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/13/2022] [Accepted: 08/03/2022] [Indexed: 01/25/2023] Open
Abstract
Previous studies have demonstrated that a high-protein diet leads to increased atherosclerosis in mice, and that this adverse effect is caused by activation of macrophage mTORC1 signaling. Here, we provide a detailed protocol for the evaluation of diet-induced mTORC1 signaling in plaque macrophages in atherosclerosis-prone apolipoprotein E (ApoE) knockout (KO) mice. This protocol includes flow cytometry and immunofluorescence analysis of atherosclerotic macrophages that can be used to study the atherogenic potential of a variety of mTORC1 modulators. For complete details on the use and execution of this protocol, please refer to Zhang et al. (2020).
Collapse
Affiliation(s)
- Xiangyu Zhang
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Jeremiah Stitham
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | | | - Se-Jin Jeong
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Arick Park
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Yu-Sheng Yeh
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Divya Kapoor
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA; John Cochran VA Medical Center, St. Louis, MO, USA
| | - Bettina Mittendorfer
- Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Babak Razani
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA; Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA; John Cochran VA Medical Center, St. Louis, MO, USA.
| |
Collapse
|
48
|
Nogueira ME, Sousa Neto IV, Motta-Santos D, Cantuária APDC, Lima SMDF, Rezende TMB, Santana HADP, Petriz BA, Marqueti RDC, Almeida JA. High-protein diet associated with resistance training reduces cardiac TNF-α levels and up-regulates MMP-2 activity in rats. Arch Physiol Biochem 2022; 128:1630-1636. [PMID: 32686511 DOI: 10.1080/13813455.2020.1787456] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The consumption of high-protein diets (HPD) is associated with resistance training (RT) due to effects on metabolism. However, little is known about these effects on cardiac tissue. This study aimed to investigate effects of HPD and RT on cardiac biomarkers. 18 rats were divided into normo-protein (NPD), and HPD groups: NPD-Control, NPD-RT, HPD-Control, and HPD-RT. Interleukin-6 (IL-6), tumour necrosis factor alpha (TNF-a), nitric oxide (NO), activity of metalloproteinase-2 (MMP-2), and vascular factor (VEGF) were analysed. RT was effective in regulating body weight, increasing strength, and reducing food consumption (p < .05). HPD induces higher levels of interleukin 6 (p = .0169), and lowers NO (p < .0001). When associated with RT, the HPD decreases levels of tumour necrosis factor alpha, while enhances NO, and MMP activity (p < .05). The association of RT with HDP decreases inflammatory parameters and indicates an enhancement in the molecular parameters of cardiac tissue.
Collapse
Affiliation(s)
- Murilo Esteves Nogueira
- Graduate Program in Health and Development in the Midwest Region, Faculty of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Brazil
| | - Ivo Vieira Sousa Neto
- Graduate Program of Sciences and Technology of Health, University of Brasilia, Distrito Federal, Brazil
| | - Daisy Motta-Santos
- Sports Science Postgraduate Program, Sports Department, EEFFTO, UFMG, Belo Horizonte, Brazil
| | - Ana Paula de Castro Cantuária
- Graduate Program in Genomics Science and Biotechnology, Catholic University of Brasilia, Brasília, Brazil
- Post graduation of Health Sciences, University of Brasilia, Distrito Federal, Brazil
| | - Stella Maris de Freitas Lima
- Graduate Program in Genomics Science and Biotechnology, Catholic University of Brasilia, Brasília, Brazil
- Dental course, Catholic University of Brasilia, Brasília, Brazil
| | - Taia Maria Berto Rezende
- Graduate Program in Genomics Science and Biotechnology, Catholic University of Brasilia, Brasília, Brazil
- Post graduation of Health Sciences, University of Brasilia, Distrito Federal, Brazil
- Dental course, Catholic University of Brasilia, Brasília, Brazil
| | - Hugo Alexandre de Paula Santana
- Research in Exercise and Nutrition in Health and Sports Performance - PENSARE, Graduate Program in Movement Sciences, Federal University of Mato Grosso do Sul, Campo Grande, Brazil
| | - Bernardo Assis Petriz
- Graduate Program in Genomics Science and Biotechnology, Catholic University of Brasilia, Brasília, Brazil
- Laboratory of Molecular Exercise Physiology, University Center - UDF, Brasília, Brazil
- Graduate Program in Health Promotion, University of Franca - UNIFRAN, Franca, Brazil
| | - Rita de Cássia Marqueti
- Graduate Program of Sciences and Technology of Health, University of Brasilia, Distrito Federal, Brazil
- Graduate Program of Rehabilitation Sciences, University of Brasilia, Distrito Federal, Brazil
| | - Jeeser Alves Almeida
- Graduate Program in Health and Development in the Midwest Region, Faculty of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Brazil
- Research in Exercise and Nutrition in Health and Sports Performance - PENSARE, Graduate Program in Movement Sciences, Federal University of Mato Grosso do Sul, Campo Grande, Brazil
| |
Collapse
|
49
|
Lu YP, Wu HW, Zhu T, Li XT, Zuo J, Hasan AA, Reichetzeder C, Delic D, Yard B, Klein T, Krämer BK, Zhang ZY, Wang XH, Yin LH, Dai Y, Zheng ZH, Hocher B. Empagliflozin reduces kidney fibrosis and improves kidney function by alternative macrophage activation in rats with 5/6-nephrectomy. Biomed Pharmacother 2022; 156:113947. [DOI: 10.1016/j.biopha.2022.113947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/20/2022] [Accepted: 10/28/2022] [Indexed: 11/02/2022] Open
|
50
|
Li YJ, Jin X, Li D, Lu J, Zhang XN, Yang SJ, Zhao YX, Wu M. New insights into vascular aging: Emerging role of mitochondria function. Biomed Pharmacother 2022; 156:113954. [DOI: 10.1016/j.biopha.2022.113954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022] Open
|