1
|
Hou Z, Wang Z, Zhang J, Liu Y, Luo Z. Effects of cannabidiol on AMPKα2 /HIF-1α/BNIP3/NIX signaling pathway in skeletal muscle injury. Front Pharmacol 2024; 15:1450513. [PMID: 39502531 PMCID: PMC11536269 DOI: 10.3389/fphar.2024.1450513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/02/2024] [Indexed: 11/08/2024] Open
Abstract
Cannabidiol: (CBD) is a non-psychoactive natural active ingredient from cannabis plant, which has many pharmacological effects, including neuroprotection, antiemetic, anti-inflammatory and anti-skeletal muscle injury. However, the mechanism of its effect on skeletal muscle injury still needs further research. In order to seek a scientifically effective way to combat skeletal muscle injury during exercise, we used healthy SD rats to establish an exercise-induced skeletal muscle injury model by treadmill training, and systematically investigated the effects and mechanisms of CBD, a natural compound in the traditional Chinese medicine Cannabis sativa L., on combating skeletal muscle injury during exercise. CBD effectively improved the fracture of skeletal muscle tissue and reduced the degree of inflammatory cell infiltration. Biochemical indexes such as CK, T, Cor, LDH, SOD, MDA, and GSH-Px in serum of rats returned to normal. Combining transcriptome and network analysis results, CBD may play a protective role in exercise-induced skeletal muscle injury through HIF-1 signaling pathway. The experimental results implied that CBD could down-regulate the expression of IL-6, NF-κB, TNF-α, Keap1, AMPKα2, HIF-1α, BNIP3 and NIX, and raised the protein expression of IL-10, Nrf2 and HO-1. These results indicate that the protective effect of CBD on exercise-induced skeletal muscle injury may be related to the inhibition of oxidative stress and inflammation, thus inhibiting skeletal muscle injury through AMPKα2/HIF-1α/BNIP3/NIX signal pathways.
Collapse
Affiliation(s)
| | - Zhifang Wang
- College of physical education, Yanshan University, Qinhuangdao, China
| | - Jun Zhang
- College of physical education, Yanshan University, Qinhuangdao, China
| | - Yunen Liu
- Shenyang Medical College, Shenyang, China
| | | |
Collapse
|
2
|
Wrona MV, Ghosh R, Coll K, Chun C, Yousefzadeh MJ. The 3 I's of immunity and aging: immunosenescence, inflammaging, and immune resilience. FRONTIERS IN AGING 2024; 5:1490302. [PMID: 39478807 PMCID: PMC11521913 DOI: 10.3389/fragi.2024.1490302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
As we age, our immune system's ability to effectively respond to pathogens declines, a phenomenon known as immunosenescence. This age-related deterioration affects both innate and adaptive immunity, compromising immune function and leading to chronic inflammation that accelerates aging. Immunosenescence is characterized by alterations in immune cell populations and impaired functionality, resulting in increased susceptibility to infections, diminished vaccine efficacy, and higher prevalence of age-related diseases. Chronic low-grade inflammation further exacerbates these issues, contributing to a decline in overall health and resilience. This review delves into the characteristics of immunosenescence and examines the various intrinsic and extrinsic factors contributing to immune aging and how the hallmarks of aging and cell fates can play a crucial role in this process. Additionally, it discusses the impact of sex, age, social determinants, and gut microbiota health on immune aging, illustrating the complex interplay of these factors in altering immune function. Furthermore, the concept of immune resilience is explored, focusing on the metrics for assessing immune health and identifying strategies to enhance immune function. These strategies include lifestyle interventions such as diet, regular physical activity, stress management, and the use of gerotherapeutics and other approaches. Understanding and mitigating the effects of immunosenescence are crucial for developing interventions that support robust immune responses in aged individuals.
Collapse
Affiliation(s)
- Marianna V. Wrona
- Columbia University in the City of New York, New York, NY, United States
| | - Rituparna Ghosh
- Columbia Center for Human Longevity, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Kaitlyn Coll
- Florida International University, Miami, FL, United States
| | - Connor Chun
- Bronx High School of Science, New York, NY, United States
| | - Matthew J. Yousefzadeh
- Columbia University in the City of New York, New York, NY, United States
- Columbia Center for Human Longevity, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Department of Medicine, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
3
|
Romero M, Gelsomini A, Miller K, Suresh D, Thaller S, Frasca D. In Vitro Treatment with Metformin Significantly Reduces Senescent B Cells Present in the Adipose Tissue of People with Obesity. J Nutr 2024:S0022-3166(24)01080-0. [PMID: 39389182 DOI: 10.1016/j.tjnut.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Our previous work has shown that senescent B cells accumulate in the human adipose tissue (AT) from people with obesity, where they express transcripts for markers associated with the senescence-associated secretory phenotype (SASP) and secrete multiple inflammatory mediators. These functions of AT-derived B cells are metabolically supported. OBJECTIVES To show that Metformin (MET), a widely used hypoglycemic and antidiabetic drug, is able at least in vitro to decrease frequencies, secretory profile, and metabolic requirements of senescent B cells isolated from the AT from people with obesity. METHODS We recruited adult females with obesity (n = 8, age 40 ± 2 y, BMI range: 33-42) undergoing breast reduction surgery, who donated their discarded subcutaneous AT. B cells from stromal vascular fractions isolated after collagenase digestion of the AT were evaluated after in vitro incubation with MET (1 mM × 106 B cells) or with a medium for the following measures. The expression of transcripts for SASP-associated markers (p16INK4a and p21CIP1/WAF1) measured by quantitative polymerase chain reaction (qPCR); secretion of inflammatory cytokines (TNF-α, IL-6, IFN-γ and IL-17A) measured by a Cytometric Bead Array); metabolic characteristics as identified by a glycolytic test and Seahorse technology, and by the expression of transcripts for glucose transporters and metabolic enzymes involved in glucose metabolic pathways, measured by qPCR. To examine differences between MET-treated compared with untreated groups, paired Student's t tests (two-tailed) were employed. RESULTS MET in vitro was able to reduce frequencies and numbers of senescent B cells, as identified by staining with β-galactosidase, as well as the secretion of inflammatory cytokines, the expression of transcripts for SASP, and metabolic markers that support intrinsic B cell inflammation. CONCLUSIONS Our results provide evidence to support the beneficial effects of MET in reducing AT-related inflammation through its effects on senescent B cells.
Collapse
Affiliation(s)
| | | | | | | | - Seth Thaller
- DeWitt Daughtry Family Department of Surgery, Division of Plastic and Reconstructive Surgery
| | - Daniela Frasca
- Department of Microbiology and Immunology; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States.
| |
Collapse
|
4
|
Nejabat M, Samie A, Khojastehnezhad A, Hadizadeh F, Ramezani M, Alibolandi M, Abnous K, Taghdisi SM, Siaj M. Stimuli-Responsive Covalent Organic Frameworks for Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:51837-51859. [PMID: 39163539 DOI: 10.1021/acsami.4c07040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Chemotherapy as a common anticancer therapeutic modality is often challenged by various obstacles such as poor stability, low solubility, and severe side effects of chemotherapeutic agents as well as multidrug resistance of cancerous cells. Nanoparticles in the role of carriers for chemotherapeutic drugs and platforms for combining different therapeutic approaches have effectively participated in overcoming such drawbacks. In particular, nanoparticles able to induce their therapeutic effect in response to specific stimuli like tumor microenvironment characteristics (e.g., hypoxia, acidic pH, high levels of glutathione, and overexpressed hydrogen peroxide) or extrinsic stimulus of laser light bring about more precise and selective treatments. Among them, nanostructures of covalent organic frameworks (COFs) have drawn great interest in biomedical fields during recent years. Possessing large surface area, high porosity, structural stability, and customizable architecture, these biocompatible porous crystalline polymers properly translate to promising platforms for drug delivery and induction of combination therapies. With the focus on stimuli-responsive characteristics of nanoscale COFs, this study aims to propose an overview of their potentiality in cancer treatment on the basis of chemotherapy alone or in combination with sonodynamic, chemodynamic, photodynamic, and photothermal therapies.
Collapse
Affiliation(s)
- Masoud Nejabat
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Ali Samie
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Amir Khojastehnezhad
- Department of Chemistry, University of Quebec at Montreal, Montreal, Quebec H3C 3P8, Canada
| | - Farzin Hadizadeh
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Mohamed Siaj
- Department of Chemistry, University of Quebec at Montreal, Montreal, Quebec H3C 3P8, Canada
| |
Collapse
|
5
|
Yin L, Niu Y, Zheng X, Chu J, Ma T. d-galactose causes embryonic development arrest and placental development disorders in mice by increasing ROS and inhibiting SIRT1/FOXO3a axis. Placenta 2024; 150:52-61. [PMID: 38593636 DOI: 10.1016/j.placenta.2024.04.003] [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: 01/13/2024] [Revised: 03/17/2024] [Accepted: 04/05/2024] [Indexed: 04/11/2024]
Abstract
INTRODUCTION Does an elevation in d-Galactose (D-Gal) levels within the body contribute to abnormal embryonic development and placental dysfunction during pregnancy? METHODS Mouse embryos were cultivated to the blastocyst stage under varying concentrations of D-Gal. The blastocyst formation rate was measured, and the levels of reactive oxygen species (ROS), sirtuin 1 (SIRT1), and forkhead box O3a (FOXO3a) in blastocysts were assessed. Mice were intraperitoneally injected with either saline or D-Gal with or without SRT1720. On the 14th day of pregnancy, the fetal absorption rate and placental weight were recorded. Placental levels of superoxide dismutase (SOD) and malondialdehyde (MDA) were determined. The expression of senescence-related factors, such as senescence-associated β-galactosidase (SA-β-gal) in the placenta was examined, and the expression of placental SIRT1, FOXO3a and p21 was evaluated by immunohistochemistry and Western blotting. RESULTS D-Gal adversely affects early embryonic development in vitro, resulting in a decreased blastocyst formation rate. Furthermore, D-Gal downregulates SIRT1 and FOXO3a while increasing ROS levels in blastocysts. Concurrently, D-Gal induces placental dysfunction, characterized by an elevated fetal absorption rate, reduced placental weight, diminished SOD activity, and increased MDA content. The senescence-related factor SA-β-gal was detected in the placenta, along with altered expression of placental SIRT1, FOXO3a, and p21. The SIRT1 agonist SRT1720 mitigated this damage by increasing SIRT1 and FOXO3a expression. DISCUSSION The inhibition of early embryonic development and placental dysfunction induced by D-Gal may be attributed to the dysregulation of SIRT1. Activating SIRT1 emerges as a potentially effective strategy for alleviating the adverse effects of D-Gal exposure.
Collapse
Affiliation(s)
- Lanlan Yin
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yanru Niu
- Laboratory of Bone Science, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xiudan Zheng
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jiaqi Chu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Tianzhong Ma
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| |
Collapse
|
6
|
Xu LL, Xie JQ, Shen JJ, Ying MD, Chen XZ. Neuron-derived exosomes mediate sevoflurane-induced neurotoxicity in neonatal mice via transferring lncRNA Gas5 and promoting M1 polarization of microglia. Acta Pharmacol Sin 2024; 45:298-311. [PMID: 37803140 PMCID: PMC10789735 DOI: 10.1038/s41401-023-01173-9] [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: 05/16/2023] [Accepted: 09/17/2023] [Indexed: 10/08/2023] Open
Abstract
Sevoflurane exposure during rapid brain development induces neuronal apoptosis and causes memory and cognitive deficits in neonatal mice. Exosomes that transfer genetic materials including long non-coding RNAs (lncRNAs) between cells play a critical role in intercellular communication. However, the lncRNAs found in exosomes derived from neurons treated with sevoflurane and their potential role in promoting neurotoxicity remain unknown. In this study, we investigated the role of cross-talk of newborn mouse neurons with microglial cells in sevoflurane-induced neurotoxicity. Mouse hippocampal neuronal HT22 cells were exposed to sevoflurane, and then co-cultured with BV2 microglial cells. We showed that sevoflurane treatment markedly increased the expression of the lncRNA growth arrest-specific 5 (Gas5) in neuron-derived extracellular vesicles, which inhibited neuronal proliferation and induced neuronal apoptosis by promoting M1 polarization of microglia and the release of inflammatory cytokines. We further revealed that the exosomal lncRNA Gas5 significantly upregulated Foxo3 as a competitive endogenous RNA of miR-212-3p in BV2 cells, and activated the NF-κB pathway to promote M1 microglial polarization and the secretion of inflammatory cytokines, thereby exacerbating neuronal damage. In neonatal mice, intracranial injection of the exosomes derived from sevoflurane-treated neurons into the bilateral hippocampi significantly increased the proportion of M1 microglia, inhibited neuronal proliferation and promoted apoptosis, ultimately leading to neurotoxicity. Similar results were observed in vitro in BV2 cells treated with the CM from HT22 cells after sevoflurane exposure. We conclude that sevoflurane induces the transfer of lncRNA Gas5-containing exosomes from neurons, which in turn regulates the M1 polarization of microglia and contributes to neurotoxicity. Thus, modulating the expression of lncRNA Gas5 or the secretion of exosomes could be a strategy for addressing sevoflurane-induced neurotoxicity.
Collapse
Affiliation(s)
- Li-Li Xu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Jia-Qian Xie
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jian-Jun Shen
- Department of Anesthesia, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Mei-Dan Ying
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China.
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Xin-Zhong Chen
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
7
|
Kang BG, Shende M, Inci G, Park SH, Jung JS, Kim SB, Kim JH, Mo YW, Seo JH, Feng JH, Kim SC, Lim SS, Suh HW, Lee JY. Combination of metformin/efavirenz/fluoxetine exhibits profound anticancer activity via a cancer cell-specific ROS amplification. Cancer Biol Ther 2023; 24:20-32. [PMID: 36588385 PMCID: PMC9809943 DOI: 10.1080/15384047.2022.2161803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The possible anticancer activity of combination (M + E + F) of metformin (M), efavirenz (E), and fluoxetine (F) was investigated in normal HDF cells and HCT116 human colon cancer cells. Metformin increased cellular FOXO3a, p-FOXO3a, AMPK, p-AMPK, and MnSOD levels in HDFs but not in HCT116 cells. Cellular ATP level was decreased only in HDFs by metformin. Metformin increased ROS level only in HCT116 cells. Transfection of si-FOXO3a into HCT116 reversed the metformin-induced cellular ROS induction, indicating that FOXO3a/MnSOD is the key regulator for cellular ROS level. Viability readout with M, E, and F alone decreased slightly, but the combination of three drugs dramatically decreased cell survival in HCT116, A549, and SK-Hep-1 cancer cells but not in HDF cells. ROS levels in HCT116 cells were massively increased by M + E + F combination, but not in HDF cells. Cell cycle analysis showed that of M + E + F combination caused cell death only in HCT116 cells. The combination of M + E + F reduced synergistically mitochondrial membrane potential and mitochondrial electron transport chain complex I and III activities in HCT116 cells when compared with individual treatments. Western blot analysis indicated that DNA damage, apoptosis, autophagy, and necroptosis-realated factors increased in M + E + F-treated HCT116 cells. Oral administration with M + E + F combination for 3 weeks caused dramatic reductions in tumor volume and weight in HCT116 xenograft model of nude mice when compared with untreated ones. Our results suggest that M + E + F have profound anticancer activity both in vitro and in vivo via a cancer cell-specific ROS amplification (CASRA) through ROS-induced DNA damage, apoptosis, autophagy, and necroptosis.
Collapse
Affiliation(s)
- Beom-Goo Kang
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Madhuri Shende
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Gozde Inci
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | | | | | | | | | | | | | | | - Sung-Chan Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Soon Sung Lim
- Department of Food Science and Nutrition, College of Natural Science, Hallym University, Chuncheon, Republic of Korea
| | - Hong-Won Suh
- FrontBio Inc, Gangwon-do, Republic of Korea,Department of Pharmacology, Institute of Natural Medicine, Hallym University, Chuncheon, Republic of Korea,Hong-Won Suh Department of Biochemistry, College of Medicine, Hallym University, 1 Hallymdeahak-gil, Chuncheon24252, Republic of Korea
| | - Jae-Yong Lee
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon, Republic of Korea,FrontBio Inc, Gangwon-do, Republic of Korea,CONTACT Jae-Yong Lee
| |
Collapse
|
8
|
Isop LM, Neculau AE, Necula RD, Kakucs C, Moga MA, Dima L. Metformin: The Winding Path from Understanding Its Molecular Mechanisms to Proving Therapeutic Benefits in Neurodegenerative Disorders. Pharmaceuticals (Basel) 2023; 16:1714. [PMID: 38139841 PMCID: PMC10748332 DOI: 10.3390/ph16121714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/25/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Metformin, a widely prescribed medication for type 2 diabetes, has garnered increasing attention for its potential neuroprotective properties due to the growing demand for treatments for Alzheimer's, Parkinson's, and motor neuron diseases. This review synthesizes experimental and clinical studies on metformin's mechanisms of action and potential therapeutic benefits for neurodegenerative disorders. A comprehensive search of electronic databases, including PubMed, MEDLINE, Embase, and Cochrane library, focused on key phrases such as "metformin", "neuroprotection", and "neurodegenerative diseases", with data up to September 2023. Recent research on metformin's glucoregulatory mechanisms reveals new molecular targets, including the activation of the LKB1-AMPK signaling pathway, which is crucial for chronic administration of metformin. The pleiotropic impact may involve other stress kinases that are acutely activated. The precise role of respiratory chain complexes (I and IV), of the mitochondrial targets, or of the lysosomes in metformin effects remains to be established by further research. Research on extrahepatic targets like the gut and microbiota, as well as its antioxidant and immunomodulatory properties, is crucial for understanding neurodegenerative disorders. Experimental data on animal models shows promising results, but clinical studies are inconclusive. Understanding the molecular targets and mechanisms of its effects could help design clinical trials to explore and, hopefully, prove its therapeutic effects in neurodegenerative conditions.
Collapse
Affiliation(s)
- Laura Mihaela Isop
- Department of Fundamental, Prophylactic and Clinical Sciences, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania; (L.M.I.)
| | - Andrea Elena Neculau
- Department of Fundamental, Prophylactic and Clinical Sciences, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania; (L.M.I.)
| | - Radu Dan Necula
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania
| | - Cristian Kakucs
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania
| | - Marius Alexandru Moga
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania
| | - Lorena Dima
- Department of Fundamental, Prophylactic and Clinical Sciences, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania; (L.M.I.)
| |
Collapse
|
9
|
Martin DE, Cadar AN, Bartley JM. Old drug, new tricks: the utility of metformin in infection and vaccination responses to influenza and SARS-CoV-2 in older adults. FRONTIERS IN AGING 2023; 4:1272336. [PMID: 37886013 PMCID: PMC10598609 DOI: 10.3389/fragi.2023.1272336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023]
Abstract
In the face of global pathogens such as influenza (flu) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), strategies beyond standard vaccines and virus-specific treatments are critically needed for older populations who are more susceptible to severe disease and death from these infections due to age-related immune dysregulation. Thus, complimentary therapeutics are needed to address the increased risk of complications and death in older adults. Metformin, an FDA approved diabetes drug, is an attractive therapeutic candidate to improve immune defenses and resilience in older adults facing viral challenge. Metformin is already a candidate anti-aging drug, but its benefits have potential to span beyond this and improve specific immune responses. Metformin can target multiple aging hallmarks as well as directly impact innate and adaptive immune cell subsets. Both retrospective and prospective studies have demonstrated metformin's efficacy in improving outcomes after SARS-CoV-2 or flu infections. Moreover, evidence from clinical trials has also suggested that metformin treatment can improve vaccination responses. In totality, these findings suggest that metformin can improve age-related declines in immunological resilience. Strategies to improve outcomes after infection or improve vaccine-induced protection are invaluable for older adults. Moreover, the ability to repurpose an already FDA approved drug has significant advantages in terms of necessary time and resources. Thus, metformin has great potential as a therapeutic to improve age-related immune dysregulation during flu and SARS-CoV-2 infections and should be further explored to confirm its ability to improve overall immunological resilience in older adults.
Collapse
|
10
|
Francisco JT, Holt AW, Bullock MT, Williams MD, Poovey CE, Holland NA, Brault JJ, Tulis DA. FoxO3 normalizes Smad3-induced arterial smooth muscle cell growth. Front Physiol 2023; 14:1136998. [PMID: 37693008 PMCID: PMC10483145 DOI: 10.3389/fphys.2023.1136998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023] Open
Abstract
Transition of arterial smooth muscle (ASM) from a quiescent, contractile state to a growth-promoting state is a hallmark of cardiovascular disease (CVD), a leading cause of death and disability in the United States and worldwide. While many individual signals have been identified as important mechanisms in this phenotypic conversion, the combined impact of the transcription factors Smad3 and FoxO3 in ASM growth is not known. The purpose of this study was to determine that a coordinated, phosphorylation-specific relationship exists between Smad3 and FoxO3 in the control of ASM cell growth. Using a rat in vivo arterial injury model and rat primary ASM cell lysates and fractions, validated low and high serum in vitro models of respective quiescent and growth states, and adenoviral (Ad-) gene delivery for overexpression (OE) of individual and combined Smad3 and/or FoxO3, we hypothesized that FoxO3 can moderate Smad3-induced ASM cell growth. Key findings revealed unique cellular distribution of Smad3 and FoxO3 under growth conditions, with induction of both nuclear and cytosolic Smad3 yet primarily cytosolic FoxO3; Ad-Smad3 OE leading to cytosolic and nuclear expression of phosphorylated and total Smad3, with almost complete reversal of each with Ad-FoxO3 co-infection in quiescent and growth conditions; Ad-FoxO3 OE leading to enhanced cytosolic expression of phosphorylated and total FoxO3, both reduced with Ad-Smad3 co-infection in quiescent and growth conditions; Ad-FoxO3 inducing expression and activity of the ubiquitin ligase MuRF-1, which was reversed with concomitant Ad-Smad3 OE; and combined Smad3/FoxO3 OE reversing both the pro-growth impact of singular Smad3 and the cytostatic impact of singular FoxO3. A primary takeaway from these observations is the capacity of FoxO3 to reverse growth-promoting effects of Smad3 in ASM cells. Additional findings lend support for reciprocal antagonism of Smad3 on FoxO3-induced cytostasis, and these effects are dependent upon discrete phosphorylation states and cellular localization and involve MuRF-1 in the control of ASM cell growth. Lastly, results showing capacity of FoxO3 to normalize Smad3-induced ASM cell growth largely support our hypothesis, and overall findings provide evidence for utility of Smad3 and/or FoxO3 as potential therapeutic targets against abnormal ASM growth in the context of CVD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - David A. Tulis
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| |
Collapse
|
11
|
Hieber C, Grabbe S, Bros M. Counteracting Immunosenescence-Which Therapeutic Strategies Are Promising? Biomolecules 2023; 13:1085. [PMID: 37509121 PMCID: PMC10377144 DOI: 10.3390/biom13071085] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Aging attenuates the overall responsiveness of the immune system to eradicate pathogens. The increased production of pro-inflammatory cytokines by innate immune cells under basal conditions, termed inflammaging, contributes to impaired innate immune responsiveness towards pathogen-mediated stimulation and limits antigen-presenting activity. Adaptive immune responses are attenuated as well due to lowered numbers of naïve lymphocytes and their impaired responsiveness towards antigen-specific stimulation. Additionally, the numbers of immunoregulatory cell types, comprising regulatory T cells and myeloid-derived suppressor cells, that inhibit the activity of innate and adaptive immune cells are elevated. This review aims to summarize our knowledge on the cellular and molecular causes of immunosenescence while also taking into account senescence effects that constitute immune evasion mechanisms in the case of chronic viral infections and cancer. For tumor therapy numerous nanoformulated drugs have been developed to overcome poor solubility of compounds and to enable cell-directed delivery in order to restore immune functions, e.g., by addressing dysregulated signaling pathways. Further, nanovaccines which efficiently address antigen-presenting cells to mount sustained anti-tumor immune responses have been clinically evaluated. Further, senolytics that selectively deplete senescent cells are being tested in a number of clinical trials. Here we discuss the potential use of such drugs to improve anti-aging therapy.
Collapse
Affiliation(s)
- Christoph Hieber
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
12
|
Yang L, He S, Ling L, Wang F, Xu L, Fang L, Wu F, Zhou S, Yang F, Wei H, Yu D. Crosstalk between miR-144/451 and Nrf2 during Recovery from Acute Hemolytic Anemia. Genes (Basel) 2023; 14:genes14051011. [PMID: 37239374 DOI: 10.3390/genes14051011] [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: 02/16/2023] [Revised: 04/04/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
miR-144/451 and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) regulate two antioxidative systems that have been identified to maintain redox homeostasis in erythroid cells by removing excess reactive oxygen species (ROS). Whether these two genes coordinate to affect ROS scavenging and the anemic phenotype, or which gene is more important for recovery from acute anemia, has not been explored. To address these questions, we crossed miR-144/451 knockout (KO) and Nrf2 KO mice and examined the phenotype change in the animals as well as the ROS levels in erythroid cells either at baseline or under stress condition. Several discoveries were made in this study. First, Nrf2/miR-144/451 double-KO mice unexpectedly exhibit similar anemic phenotypes as miR-144/451 single-KO mice during stable erythropoiesis, although compound mutations of miR-144/451 and Nrf2 lead to higher ROS levels in erythrocytes than single gene mutations. Second, Nrf2/miR-144/451 double-mutant mice exhibit more dramatic reticulocytosis than miR-144/451 or Nrf2 single-KO mice during days 3 to 7 after inducing acute hemolytic anemia using phenylhydrazine (PHZ), indicating a synergistic effect of miR-144/451 and Nrf2 on PHZ-induced stress erythropoiesis. However, the coordination does not persist during the whole recovery stage of PHZ-induced anemia; instead, Nrf2/miR-144/451 double-KO mice follow a recovery pattern similar to miR-144/451 single-KO mice during the remaining period of erythropoiesis. Third, the complete recovery from PHZ-induced acute anemia in miR-144/451 KO mice takes longer than in Nrf2 KO mice. Our findings demonstrate that complicated crosstalk between miR-144/451 and Nrf2 does exist and the crosstalk of these two antioxidant systems is development-stage-dependent. Our findings also demonstrate that miRNA deficiency could result in a more profound defect of erythropoiesis than dysfunctional transcription factors.
Collapse
Affiliation(s)
- Lei Yang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou 225009, China
| | - Sheng He
- Guangxi Key Laboratory of Birth Defects Research and Prevention, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Zhuang Autonomous Region Women and Children Care Hospital, Nanning 530000, China
| | - Ling Ling
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou 225009, China
| | - Fangfang Wang
- Department of Hematology, Yangzhou University Clinical Medical College, Yangzhou 225001, China
| | - Lei Xu
- Central Laboratory, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225003, China
| | - Lei Fang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou 225009, China
| | - Fan Wu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou 225009, China
| | - Shuting Zhou
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou 225009, China
| | - Fan Yang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou 225009, China
| | - Hongwei Wei
- Guangxi Key Laboratory of Birth Defects Research and Prevention, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Zhuang Autonomous Region Women and Children Care Hospital, Nanning 530000, China
| | - Duonan Yu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou 225009, China
- Guangxi Key Laboratory of Birth Defects Research and Prevention, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Zhuang Autonomous Region Women and Children Care Hospital, Nanning 530000, China
| |
Collapse
|
13
|
Makrutzki-Zlotek K, Escher F, Karadeniz Z, Aleshcheva G, Pietsch H, Küchler K, Schultheiss HP, Heidecker B, Poller W, Landmesser U, Scheibenbogen C, Thevathasan T, Skurk C. FOXO3A acts as immune response modulator in human virus-negative inflammatory cardiomyopathy. HEART (BRITISH CARDIAC SOCIETY) 2023; 109:846-856. [PMID: 36702542 DOI: 10.1136/heartjnl-2022-321732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 01/02/2023] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Inflammatory cardiomyopathy is characterised by inflammatory infiltrates leading to cardiac injury, left ventricular (LV) dilatation and reduced LV ejection fraction (LVEF). Several viral pathogens and autoimmune phenomena may cause cardiac inflammation.The effects of the gain of function FOXO3A single-nucleotide polymorphism (SNP) rs12212067 on inflammation and outcome were studied in a cohort of patients with inflammatory dilated cardiomyopathy (DCMi) in relation to cardiac viral presence. METHODS Distribution of the SNP was determined in virus-positive and virus-negative DCMi patients and in control subjects without myocardial pathology. Baseline and outcome data were compared in 221 virus-negative patients with detection of cardiac inflammation and reduced LVEF according to their carrier status of the SNP. RESULTS Distribution of SNP rs12212067 did not differ between virus-positive (n=22, 19.3%), virus-negative (n=45, 20.4 %) and control patients (n=18, 23.4 %), indicating the absence of susceptibility for viral infection or inflammation per se (p=0.199). Patients in the virus-negative DCMi group were characterised by reduced LVEF 35.5% (95% CI) 33.5 to 37.4) and increased LVEDD (LV end-diastolic diameter) 59.8 mm (95% CI 58.5 to 61.2). Within the group, SNP and non-SNP carriers had similarly impaired LVEF 39.2% (95% CI 34.3% to 44.0%) vs 34.5% (95% CI 32.4 to 36.5), p=0.083, and increased LVEDD 58.9 mm (95% CI 56.3 to 61.5) vs 60.1 mm (95% CI 58.6 to 61.6), p=0.702, respectively. The number of inflammatory infiltrates was not different in both SNP groups at baseline. Outcome after 6 months showed a significant improvement in LVEF and clinical symptoms in SNP rs12212067 carriers 50.9% (95% CI 45.4 to 56.3) versus non-SNP carriers 41.7% (95% CI 39.2 to 44.2), p≤0.01. The improvement in clinical symptoms and LVEF was associated with a significant reduction in cardiac inflammation (ΔCD45RO+ p≤0.05; ΔMac-1+ p≤0.05; ΔLFA-1+ p≤0.01; ΔCD54+ p≤0.01) in the SNP cohort versus non-SNP cohort, respectively. Subgroup analyses identified ΔMac-1+, ΔLFA-1+, ΔCD3+ and Δperforin+ as predictors for improvement in cardiac function in SNP-positive patients. CONCLUSION FOXO3A might act as modulator of the cardiac immune response, diminishing cardiac inflammation and injury in pathogen-negative DCMi.
Collapse
Affiliation(s)
- Kamila Makrutzki-Zlotek
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
| | - Felicitas Escher
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Virchow-Klinikum, Berlin, Germany.,DZHK, German Center for Cardiovascular Research, Berlin, Germany.,IKDT, Institute for Cardiac Diagnostics and Therapy, Berlin, Germany
| | - Zehra Karadeniz
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
| | - Ganna Aleshcheva
- IKDT, Institute for Cardiac Diagnostics and Therapy, Berlin, Germany
| | - Heiko Pietsch
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Virchow-Klinikum, Berlin, Germany.,IKDT, Institute for Cardiac Diagnostics and Therapy, Berlin, Germany
| | - Konstanze Küchler
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany
| | | | - Bettina Heidecker
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany.,Institute of Medical Informatics, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang Poller
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany.,Institute of Medical Informatics, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany.,Institute of Medical Informatics, Charité Universitätsmedizin Berlin, Berlin, Germany.,BIH, Berlin Institute of Health at Charité, Berlin, Germany
| | - Carmen Scheibenbogen
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin - Campus Virchow-Klinikum, Berlin, Germany
| | - Tharusan Thevathasan
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany.,DZHK, German Center for Cardiovascular Research, Berlin, Germany.,Institute of Medical Informatics, Charité Universitätsmedizin Berlin, Berlin, Germany.,BIH, Berlin Institute of Health at Charité, Berlin, Germany
| | - Carsten Skurk
- Department of Cardiology, Deutsches Herzzentrum der Charité (DHZC), Campus Benjamin Franklin, Berlin, Germany .,DZHK, German Center for Cardiovascular Research, Berlin, Germany
| |
Collapse
|
14
|
Wang X, Ren XM, He H, Li F, Liu K, Zhao F, Hu H, Zhang P, Huang B, Pan X. Cytotoxicity and pro-inflammatory effect of polystyrene nano-plastic and micro-plastic on RAW264.7 cells. Toxicology 2023; 484:153391. [PMID: 36503103 DOI: 10.1016/j.tox.2022.153391] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/25/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
Numerous studies have shown that exposure to micro- or nano-plastics led to the cell viability and function of macrophages in the intestine tissue might be one possible mechanism. This study investigated the cytotoxicity and pro-inflammatory effect of 80 nm polystyrene-nano-plastic (PS-NP) and 3 µm PS-micro-plastic (PS-MP) on mouse macrophages RAW264.7 cells. Our results showed that exposure to PS-NP or PS-MP induced apoptosis of cells at 5 or 10 μg/mL, respectively. Besides, PS-NP enhanced the secretion of inflammatory cytokines (Tumor necrosis factor-α, Interleukin-6 and Interleukin-10) with the lowest effective concentration (LOEC) of 1, 0.01, and 0.01 μg/mL, respectively. PS-MP enhanced secretion of TNF-α and IL-10 with the LOEC of 1 and 0.01 μg/mL, respectively. We further studied the possible mechanisms of the effects of PS-NP or PS-MP on RAW264.7 cells. We found they might cause cytotoxicity and inflammatory effects by producing reactive oxygen species and nitric oxide in the cells. Accordingly, our results demonstrated that PS-NP and PS-MP had cytotoxicity and pro-inflammatory effect on macrophages, which might further lead to intestinal inflammation. Moreover, we revealed that the PS-NP had more potent adverse impacts on macrophages than PS-MP.
Collapse
Affiliation(s)
- Xiaoxia Wang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Xiao-Min Ren
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China.
| | - Huan He
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Fan Li
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Kunqian Liu
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Fenqing Zhao
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Huixiang Hu
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Pingping Zhang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Bin Huang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; Yunnan Provincial Key Laboratory of Carbon Sequestration and Pollution Control in Soils, Kunming 650500, China
| | - Xuejun Pan
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; Yunnan Provincial Key Laboratory of Carbon Sequestration and Pollution Control in Soils, Kunming 650500, China.
| |
Collapse
|
15
|
Xu L, Zhang X, Zhao Y, Gang X, Zhou T, Han J, Cao Y, Qi B, Song S, Wang X, Liang Y. Metformin protects trabecular meshwork against oxidative injury via activating integrin/ROCK signals. eLife 2023; 12:81198. [PMID: 36598818 PMCID: PMC9812404 DOI: 10.7554/elife.81198] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 12/09/2022] [Indexed: 01/05/2023] Open
Abstract
This study aimed to investigate the protective effect of metformin on trabecular meshwork (TM) and explore its molecular mechanisms in vivo and in vitro. Ocular hypertension (OHT) mouse models were induced with dexamethasone and further treated with metformin to determine the intraocular pressure (IOP)-lowering effect. Cultured human TM cells (HTMCs) were pre-stimulated with tert-butyl hydroperoxide (tBHP) to induce oxidative damage and then supplemented with metformin for another 24 hr. The expression of fibrotic markers and integrin/Rho-associated kinase (ROCK) signals, including α-smooth muscle actin (α-SMA), transforming growth factor-β (TGF-β), fibronectin, integrin beta 1, ROCK 1/2, AMP-activated protein kinase, myosin light chain 1, and F-actin were determined by western blotting and immunofluorescence. Reactive oxygen species (ROS) content was analysed using flow cytometry. Transmission electron microscopy was performed to observe microfilaments in HTMCs. It showed that metformin administration reduced the elevated IOP and alleviated the fibrotic activity of aqueous humour outflow in OHT models. Additionally, metformin rearranged the disordered cytoskeleton in the TM both in vivo and in vitro and significantly inhibited ROS production and activated integrin/ROCK signalling induced by tBHP in HTMCs. These results indicated that metformin reduced the elevated IOP in steroid-induced OHT mouse models and exerted its protective effects against oxidative injury by regulating cytoskeleton remodelling through the integrin/ROCK pathway. This study provides new insights into metformin use and preclinical evidence for the potential treatment of primary open-angle glaucoma.
Collapse
Affiliation(s)
- Lijuan Xu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, WenzhouZhejiangChina
| | - Xinyao Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, WenzhouZhejiangChina
| | - Yin Zhao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, WenzhouZhejiangChina
| | - Xiaorui Gang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, WenzhouZhejiangChina
| | - Tao Zhou
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, WenzhouZhejiangChina
| | - Jialing Han
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, WenzhouZhejiangChina
| | - Yang Cao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, WenzhouZhejiangChina
| | - Binyan Qi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, WenzhouZhejiangChina
| | - Shuning Song
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, WenzhouZhejiangChina
| | - Xiaojie Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, WenzhouZhejiangChina
| | - Yuanbo Liang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, WenzhouZhejiangChina
| |
Collapse
|
16
|
Li K, Romero M, Cañardo M, Garcia D, Diaz A, Blomberg BB, Frasca D. B cells from old mice induce the generation of inflammatory T cells through metabolic pathways. Mech Ageing Dev 2023; 209:111742. [PMID: 36309082 DOI: 10.1016/j.mad.2022.111742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022]
Abstract
We have measured the capacity of B cells from young and old mice to induce the differentiation of naïve CD4 + T cells from young mice into pro-inflammatory subsets. We found that only B cells from old mice are inflammatory and induce in vitro secretion of the pro-inflammatory cytokines IL-17A and IFN-γ by T cells. In co-culture experiments, B cells from old mice showed a strong helper function on T cells from young mice, making them pro-inflammatory, and this effect is regulated by metabolic pathways, mainly anaerobic glycolysis, leading to increased RNA expression of the enzyme lactate dehydrogenase (LDHA) and increased secretion of lactate. These results have indicated that lactate is a crucial player of the B cell-induced polarization of T cells. When we measured the effects of lactate on isolated CD4 + T cells from young mice, we found that lactate increases RNA expression of LDHA, secretion of pro-inflammatory cytokines and NF-kB activation. Moreover, lactate effects in culture can be abrogated in the presence of the specific inhibitor of LDHA, FX11. These results altogether may have relevant clinical implications and suggest novel targets for therapeutic interventions in patients with inflammatory conditions and diseases.
Collapse
Affiliation(s)
- Kevin Li
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA
| | - Maria Romero
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA
| | - Macarena Cañardo
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA
| | - Denisse Garcia
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA
| | - Alain Diaz
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA; Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine, Miami, FL USA
| | - Daniela Frasca
- Department of Microbiology and Immunology and University of Miami Miller School of Medicine, Miami, FL USA; Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine, Miami, FL USA.
| |
Collapse
|
17
|
Mu W, Jiang Y, Liang G, Feng Y, Qu F. Metformin: A Promising Antidiabetic Medication for Cancer Treatment. Curr Drug Targets 2023; 24:41-54. [PMID: 36336804 DOI: 10.2174/1389450124666221104094918] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/18/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022]
Abstract
Metformin is a widely used drug in patients with type 2 diabetes mellitus. Metformin inhibits hepatic gluconeogenesis and increases glucose utilization in peripheral tissues. In recent years, several studies have shown that metformin is a potential therapeutic agent against cancer, alone or combined with other anticancer treatments. Metformin mainly activates the AMPK complex and regulates intracellular energy status, inhibiting the mitochondrial respiratory chain complex I and reducing the production of reactive oxygen species. Other anticancer targets of metformin are specific transcription factors inhibiting cell proliferation, promoting apoptosis and reducing drug resistance. In addition, metformin modulates tumor cells' response to anticancer treatments, favoring the activity of T cells. In diabetic patients, metformin reduces the occurrence of cancer and improves the prognosis and efficacy of anticancer treatments. In this review, we provided a comprehensive perspective of metformin as an anticancer drug.
Collapse
Affiliation(s)
- Wei Mu
- Department of Pharmacy and Clinical Pharmacy, Precision Medicine Center, 904th Hospital of PLA, 214044 Wuxi, Jiangsu, PR China
| | - Yunyun Jiang
- Department of Pharmacy and Clinical Pharmacy, Precision Medicine Center, 904th Hospital of PLA, 214044 Wuxi, Jiangsu, PR China
| | - Guoqiang Liang
- Central Laboratory, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, 215000 Suzhou, Jiangsu, PR China
| | - Yue Feng
- Department of Pharmacy and Clinical Pharmacy, Precision Medicine Center, 904th Hospital of PLA, 214044 Wuxi, Jiangsu, PR China
| | - Falin Qu
- Department of Pharmacy and Clinical Pharmacy, Precision Medicine Center, 904th Hospital of PLA, 214044 Wuxi, Jiangsu, PR China
| |
Collapse
|
18
|
Dharmaratne M, Kulkarni AS, Taherian Fard A, Mar JC. scShapes: a statistical framework for identifying distribution shapes in single-cell RNA-sequencing data. Gigascience 2022; 12:giac126. [PMID: 36691728 PMCID: PMC9871437 DOI: 10.1093/gigascience/giac126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 08/27/2022] [Accepted: 12/15/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Single-cell RNA sequencing (scRNA-seq) methods have been advantageous for quantifying cell-to-cell variation by profiling the transcriptomes of individual cells. For scRNA-seq data, variability in gene expression reflects the degree of variation in gene expression from one cell to another. Analyses that focus on cell-cell variability therefore are useful for going beyond changes based on average expression and, instead, identifying genes with homogeneous expression versus those that vary widely from cell to cell. RESULTS We present a novel statistical framework, scShapes, for identifying differential distributions in single-cell RNA-sequencing data using generalized linear models. Most approaches for differential gene expression detect shifts in the mean value. However, as single-cell data are driven by overdispersion and dropouts, moving beyond means and using distributions that can handle excess zeros is critical. scShapes quantifies gene-specific cell-to-cell variability by testing for differences in the expression distribution while flexibly adjusting for covariates if required. We demonstrate that scShapes identifies subtle variations that are independent of altered mean expression and detects biologically relevant genes that were not discovered through standard approaches. CONCLUSIONS This analysis also draws attention to genes that switch distribution shapes from a unimodal distribution to a zero-inflated distribution and raises open questions about the plausible biological mechanisms that may give rise to this, such as transcriptional bursting. Overall, the results from scShapes help to expand our understanding of the role that gene expression plays in the transcriptional regulation of a specific perturbation or cellular phenotype. Our framework scShapes is incorporated into a Bioconductor R package (https://www.bioconductor.org/packages/release/bioc/html/scShapes.html).
Collapse
Affiliation(s)
- Malindrie Dharmaratne
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Ameya S Kulkarni
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Atefeh Taherian Fard
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Jessica C Mar
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
19
|
Davoudi P, Do DN, Rathgeber B, Colombo SM, Sargolzaei M, Plastow G, Wang Z, Karimi K, Hu G, Valipour S, Miar Y. Genome-wide detection of copy number variation in American mink using whole-genome sequencing. BMC Genomics 2022; 23:649. [PMID: 36096727 PMCID: PMC9468235 DOI: 10.1186/s12864-022-08874-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/05/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Copy number variations (CNVs) represent a major source of genetic diversity and contribute to the phenotypic variation of economically important traits in livestock species. In this study, we report the first genome-wide CNV analysis of American mink using whole-genome sequence data from 100 individuals. The analyses were performed by three complementary software programs including CNVpytor, DELLY and Manta. RESULTS A total of 164,733 CNVs (144,517 deletions and 20,216 duplications) were identified representing 5378 CNV regions (CNVR) after merging overlapping CNVs, covering 47.3 Mb (1.9%) of the mink autosomal genome. Gene Ontology and KEGG pathway enrichment analyses of 1391 genes that overlapped CNVR revealed potential role of CNVs in a wide range of biological, molecular and cellular functions, e.g., pathways related to growth (regulation of actin cytoskeleton, and cAMP signaling pathways), behavior (axon guidance, circadian entrainment, and glutamatergic synapse), lipid metabolism (phospholipid binding, sphingolipid metabolism and regulation of lipolysis in adipocytes), and immune response (Wnt signaling, Fc receptor signaling, and GTPase regulator activity pathways). Furthermore, several CNVR-harbored genes associated with fur characteristics and development (MYO5A, RAB27B, FGF12, SLC7A11, EXOC2), and immune system processes (SWAP70, FYN, ORAI1, TRPM2, and FOXO3). CONCLUSIONS This study presents the first genome-wide CNV map of American mink. We identified 5378 CNVR in the mink genome and investigated genes that overlapped with CNVR. The results suggest potential links with mink behaviour as well as their possible impact on fur quality and immune response. Overall, the results provide new resources for mink genome analysis, serving as a guideline for future investigations in which genomic structural variations are present.
Collapse
Affiliation(s)
- Pourya Davoudi
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Duy Ngoc Do
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Bruce Rathgeber
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Stefanie M Colombo
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Mehdi Sargolzaei
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
- Select Sires Inc., Plain City, OH, USA
| | - Graham Plastow
- Livestock Gentec, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Zhiquan Wang
- Livestock Gentec, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Karim Karimi
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Guoyu Hu
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Shafagh Valipour
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Younes Miar
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada.
| |
Collapse
|
20
|
Zhang L, Liu X, Huang M, Wang R, Zhu W, Li Y, Shen L, Li C. Metformin Inhibits HaCaT Cell Proliferation Under Hyperlipidemia Through Reducing Reactive Oxygen Species via FOXO3 Activation. Clin Cosmet Investig Dermatol 2022; 15:1403-1413. [PMID: 35910506 PMCID: PMC9326038 DOI: 10.2147/ccid.s368845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/11/2022] [Indexed: 11/23/2022]
Abstract
Purpose Metformin (MET) has been proved to be effective for the treatment of psoriasis. The mechanisms of its action under the hyperlipidemia have yet to be fully elucidated. Here, we investigated the effect of metformin on the cell proliferation induced by hyperlipidemia and the underlying mechanism in immortalized human keratinocyte cell line (HaCat). Methods Wild-type or FOXO3 knockdown HaCat cells were treated with free fatty acids (FFA) for 10 days and then co-treated with metformin for another 4 days. Triglyceride (TG) level, cell viability, proliferation, apoptosis, antioxidant enzymes, reactive oxygen species (ROS) levels, as well as the transcription activity of FOXO3 were analyzed. Results Metformin decreased HaCaT cell proliferation and induced cell apoptosis after FFA treatment. Metformin was found to significantly increase the expressions and the activities of superoxide dismutase (SOD) as well as catalase (CAT), and reduced the reactive oxygen species (ROS) level. Metformin significantly promoted the autophagy and increase FOXO3 protein level in the nucleus under hyperlipidemia. However, all of the effects from metformin were partially blocked by FOXO3 knockdown. Conclusion This study demonstrated that under the hyperlipidemia, metformin has significant antiproliferation and proapoptosis effects by reducing ROS level as well as increasing autophagy. All of these effects from metformin were through FOXO3-dependent pathway.
Collapse
Affiliation(s)
- Li Zhang
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Xiaoling Liu
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Min Huang
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Rui Wang
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Wenwei Zhu
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Yu Li
- Department of Dermatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Lin Shen
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Chengxin Li
- Department of Dermatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| |
Collapse
|
21
|
Xie W, Chang W, Wang X, Liu F, Wang X, Yuan D, Zhang Y. Allicin Inhibits Osteosarcoma Growth by Promoting Oxidative Stress and Autophagy via the Inactivation of the lncRNA MALAT1-miR-376a-Wnt/ β-Catenin Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4857814. [PMID: 35783190 PMCID: PMC9249524 DOI: 10.1155/2022/4857814] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 12/29/2022]
Abstract
Allicin, an organic sulfur compound extracted from the bulb of Allium sativum, can potentially prevent various tumors. Our previous study found that allicin can effectively suppress the proliferation of osteosarcoma cells. However, the molecular mechanisms have not been illustrated. In this study, Saos-2 and U2OS osteosarcoma cells were used to investigate the underlying mechanisms. A series of experiments were carried out to authenticate the anticancer property of allicin. Knockdown of lncRNA MALAT1 inhibited the proliferation, invasion and migration and promoted apoptosis of osteosarcoma cells. Knockdown of miR-376a increased the proliferation, invasion, and migration and dropped apoptosis of osteosarcoma cells. Furthermore, knockdown of miR-376a reversed the influences of MALAT1 silencing in osteosarcoma cells. Based on our data, MALAT1 could downregulate the expression of miR-376a, subsequently accelerating osteosarcoma. Moreover, oxidative stress and autophagy were identified as the potential key pathway of allicin. Allicin inhibited osteosarcoma growth and promoted oxidative stress and autophagy via MALATI-miR-376a. We also found that allicin promotes oxidative stress and autophagy to inhibit osteosarcoma growth by inhibiting the Wnt/β-catenin pathway in vivo and in vitro. All data showed that allicin promotes oxidative stress and autophagy of osteosarcoma via the MALATI-miR-376a-Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Wenpeng Xie
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Wenjie Chang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Xiaole Wang
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Fei Liu
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Xu Wang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Daotong Yuan
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
| | - Yongkui Zhang
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, China
- Shandong Fupai Pharmaceutical Co., Ltd, Jinan, Shandong, 250000, China
| |
Collapse
|
22
|
How Is CYP17A1 Activity Altered in Autism? A Pilot Study to Identify Potential Pharmacological Targets. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060867. [PMID: 35743898 PMCID: PMC9225657 DOI: 10.3390/life12060867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 05/30/2022] [Accepted: 06/08/2022] [Indexed: 11/16/2022]
Abstract
Background: Increasing evidence exists that higher levels of androgens can be found in individuals with autism. Evidence yields to a susceptible role of Cytochrome P450 17A1 (CYP17A1) with its catalyzation of the two distinct types of substrate oxidation by a hydroxylase activity (17-alpha hydroxylase) and C17/20 lyase activity. However, to what extent steps are altered in affected children with autism versus healthy controls remains to be elucidated. Methods: Urine samples from 48 boys with autism (BMI 19.1 ± 0.6 kg/m2, age 14.2 ± 0.5 years) and a matched cohort of 48 healthy boys (BMI 18.6 ± 0.3 kg/m2, 14.3 ± 0.5 years) as well as 16 girls with autism (BMI 17.5 ± 0.7 kg/m2, age 13.8 ± 1.0 years) and a matched cohort of 16 healthy girls (BMI 17.2 ± 0.8 kg/m2, age 13.2 ± 0.8 years) were analyzed for steroid hormone metabolites by gas chromatography-mass spectrometry. Results: The activity of 17-alpha Hydroxylase increased by almost 50%, whereas activity of 17/20 Lyase activity increased by around 150% in affected children with autism. Furthermore, the concentration of Cortisol was higher as compared to the average increase of the three metabolites TH-Corticosterone, 5α-TH-Corticosterone and TH-11β-DH-Corticosterone, indicating, in addition, a stimulation by the CRH-ACTH system despite a higher enzymatic activity. Discussion: As it was shown that oxidative stress increases the 17/20-lyase activity via p38α, a link between higher steroid hormone levels and oxidative stress can be established. However, as glucocorticoid as well as androgen metabolites showed higher values in subjects affected with autism as compared to healthy controls, the data indicate, despite higher CYP17A1 activity, the presence of increased substrate availability in line with the Cholesterol theory of autism.
Collapse
|
23
|
Wiernsperger N, Al-Salameh A, Cariou B, Lalau JD. Protection by metformin against severe Covid-19: an in-depth mechanistic analysis. DIABETES & METABOLISM 2022; 48:101359. [PMID: 35662580 PMCID: PMC9154087 DOI: 10.1016/j.diabet.2022.101359] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 12/05/2022]
Abstract
Since the outbreak of Covid-19, several observational studies on diabetes and Covid-19 have reported a favourable association between metformin and Covid-19-related outcomes in patients with type 2 diabetes mellitus (T2DM). This is not surprising since metformin affects many of the pathophysiological mechanisms implicated in SARS-CoV-2 immune response, systemic spread and sequelae. A comparison of the multifactorial pathophysiological mechanisms of Covid-19 progression with metformin's well-known pleiotropic properties suggests that the treatment of patients with this drug might be particularly beneficial. Indeed, metformin could alleviate the cytokine storm, diminish virus entry into cells, protect against microvascular damage as well as prevent secondary fibrosis. Although our in-depth analysis covers many potential metformin mechanisms of action, we want to highlight more particularly its unique microcirculatory protective effects since worsening of Covid-19 disease clearly appears as largely due to severe defects in the structure and functioning of microvessels. Overall, these observations confirm that metformin is a unique, pleiotropic drug that targets many of Covid-19′s pathophysiology processes in a diabetes-independent manner.
Collapse
Affiliation(s)
| | - Abdallah Al-Salameh
- Department of Endocrinology, Diabetes Mellitus and Nutrition, Amiens University Hospital, Amiens, France; PériTox/UMR-I 01, University of Picardie Jules Verne, Amiens, France
| | - Bertrand Cariou
- Département d'Endocrinologie, Diabétologie et Nutrition, l'institut du thorax, Inserm, CNRS, UNIV Nantes, CHU Nantes, Hôpital Guillaume et René Laennec, 44093 Nantes Cedex 01, France
| | - Jean-Daniel Lalau
- Department of Endocrinology, Diabetes Mellitus and Nutrition, Amiens University Hospital, Amiens, France; PériTox/UMR-I 01, University of Picardie Jules Verne, Amiens, France.
| |
Collapse
|
24
|
Du MR, Gao QY, Liu CL, Bai LY, Li T, Wei FL. Exploring the Pharmacological Potential of Metformin for Neurodegenerative Diseases. Front Aging Neurosci 2022; 14:838173. [PMID: 35557834 PMCID: PMC9087341 DOI: 10.3389/fnagi.2022.838173] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/11/2022] [Indexed: 12/30/2022] Open
Abstract
Metformin, one of the first-line of hypoglycemic drugs, has cardioprotective, anti-inflammatory and anticancer activities, in addition to its proven hypoglycemic effects. Furthermore, the preventive and therapeutic potential of metformin for neurodegenerative diseases has become a topic of concern. Increasing research suggests that metformin can prevent the progression of neurodegenerative diseases. In recent years, many studies have investigated the neuroprotective effect of metformin in the treatment of neurodegenerative diseases. It has been revealed that metformin can play a neuroprotective role by regulating energy metabolism, oxidative stress, inflammatory response and protein deposition of cells, and avoiding neuronal dysfunction and neuronal death. On the contrary, some have hypothesized that metformin has a two-sided effect which may accelerate the progression of neurodegenerative diseases. In this review, the results of animal experiments and clinical studies are reviewed to discuss the application prospects of metformin in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ming-Rui Du
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Quan-You Gao
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Chen-Lin Liu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Lin-Ya Bai
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Fei-Long Wei
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
25
|
Ruskovska T, Morand C, Bonetti CI, Gebara KS, Cardozo Junior EL, Milenkovic D. Multigenomic modifications in human circulating immune cells in response to consumption of polyphenol rich extract of yerba mate ( Ilex paraguariensis A. St.-Hil.) are suggestive of cardiometabolic protective effects. Br J Nutr 2022; 129:1-60. [PMID: 35373729 DOI: 10.1017/s0007114522001027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Mate is a traditional drink obtained from the leaves of yerba mate and rich in a diversity of plant bioactive compounds including polyphenols, particularly chlorogenic acids. Studies, even though limited, suggest that consumption of mate is associated with health effects, including prevention of cardiometabolic disorders. Molecular mechanisms underlying the potential health properties are still largely unknown, especially in humans. The aim of this study was to investigate nutrigenomic effects of mate consumption and identify regulatory networks potentially mediating cardiometabolic health benefits. Healthy middle-aged men at risk for cardiovascular disease consumed a standardized mate extract or placebo for 4 weeks. Global gene expression, including protein coding and non-coding RNAs profiles were determined using microarrays. Biological function analyses were performed using integrated bioinformatic tools. Comparison of global gene expression profiles showed significant change following mate consumption with 2635 significantly differentially expressed genes, among which 6 are miRNAs and 244 are lncRNAs. Functional analyses showed that these genes are involved in regulation of cell interactions and motility, inflammation or cell signaling. Transcription factors, such as MEF2A, MYB or HNF1A, could have their activity modulated by mate consumption either by direct interaction with polyphenol metabolites or by interactions of metabolites with cell signaling proteins, like p38 or ERK1/2, that could modulate transcription factor activity and regulate expression of genes observed. Correlation analysis suggests that expression profile is inversely associated with gene expression profiles of patients with cardiometabolic disorders. Therefore, mate consumption may exert cardiometabolic protective effects by modulating gene expression towards a protective profile.
Collapse
Affiliation(s)
- Tatjana Ruskovska
- Faculty of Medical Sciences, Goce Delcev University, 2000 Stip, North Macedonia
| | - Christine Morand
- Human Nutrition Unit, Université Clermont Auvergne, INRAE, F-63003 Clermont-Ferrand, France
| | - Carla Indianara Bonetti
- Institute of Biological, Medical and Health Sciences, Universidade Paranaense, Av. Parigot de Souza, 3636 J. Prada, Toledo 85903-170, PR, Brazil
| | - Karimi Sater Gebara
- Grande Dourados University Center, UNIGRAN, R. Balbina de Matos, 2121 - J. Universitario, Dourados 79824-900, MS, Brazil
| | - Euclides Lara Cardozo Junior
- Institute of Biological, Medical and Health Sciences, Universidade Paranaense, Av. Parigot de Souza, 3636 J. Prada, Toledo 85903-170, PR, Brazil
| | - Dragan Milenkovic
- Human Nutrition Unit, Université Clermont Auvergne, INRAE, F-63003 Clermont-Ferrand, France
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| |
Collapse
|
26
|
Kulkarni AS, Aleksic S, Berger DM, Sierra F, Kuchel G, Barzilai N. Geroscience-guided repurposing of FDA-approved drugs to target aging: A proposed process and prioritization. Aging Cell 2022; 21:e13596. [PMID: 35343051 PMCID: PMC9009114 DOI: 10.1111/acel.13596] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/11/2022] [Accepted: 03/13/2022] [Indexed: 12/29/2022] Open
Abstract
Common chronic diseases represent the greatest driver of rising healthcare costs, as well as declining function, independence, and quality of life. Geroscience-guided approaches seek to delay the onset and progression of multiple chronic conditions by targeting fundamental biological pathways of aging. This approach is more likely to improve overall health and function in old age than treating individual diseases, by addressing aging the largest and mostly ignored risk factor for the leading causes of morbidity in older adults. Nevertheless, challenges in repurposing existing and moving newly discovered interventions from the bench to clinical care have impeded the progress of this potentially transformational paradigm shift. In this article, we propose the creation of a standardized process for evaluating FDA-approved medications for their geroscience potential. Criteria for systematically evaluating the existing literature that spans from animal models to human studies will permit the prioritization of efforts and financial investments for translating geroscience and allow immediate progress on the design of the next Targeting Aging with MEtformin (TAME)-like study involving such candidate gerotherapeutics.
Collapse
Affiliation(s)
- Ameya S. Kulkarni
- Institute for Aging ResearchAlbert Einstein College of MedicineBronxNew YorkUSA
- Present address:
AbbVie Inc.North ChicagoIL60064USA.
| | - Sandra Aleksic
- Department of Medicine (Endocrinology and Geriatrics)Albert Einstein College of MedicineBronxNew YorkUSA
| | - David M. Berger
- Department of Medicine (Hospital Medicine)Montefiore Medical Center and Albert Einstein College of MedicineBronxNew YorkUSA
| | - Felipe Sierra
- Centre Hospitalier Universitaire de ToulouseToulouseFrance
| | - George A. Kuchel
- UConn Center on AgingUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Nir Barzilai
- Institute for Aging ResearchAlbert Einstein College of MedicineBronxNew YorkUSA
| |
Collapse
|
27
|
Lou H, Chu L, Zhou W, Dou J, Teng X, Tan W, Zhou B. Diselenium-bridged covalent organic framework with pH/GSH/photo-triple-responsiveness for highly controlled drug release toward joint chemo/photothermal/chemodynamic cancer therapy. J Mater Chem B 2022; 10:7955-7966. [PMID: 35792081 DOI: 10.1039/d2tb01015a] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Here, a novel joint chemo/photothermal/chemodynamic therapy was developed using a pH/GSH/photo triple-responsive 2D-covalent organic framework (COFs) drug carriers for passive target treatment of tumor with extraordinarily high efficiency. The well-designed...
Collapse
Affiliation(s)
- Han Lou
- School of Pharmacy, Weifang Medical University, Weifang, 261053, Shandong, P. R. China.
- Department of Urology, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Shandong, P. R. China
| | - Lichao Chu
- Department of Anesthesiology, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, 261031, Shandong, P. R. China
| | - Wenbin Zhou
- Department of Urology, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Shandong, P. R. China
| | - Jinli Dou
- School of Pharmacy, Weifang Medical University, Weifang, 261053, Shandong, P. R. China.
| | - Xiaotong Teng
- Department of Respiratory Medicine, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, 261031, Shandong, P. R. China
| | - Wei Tan
- Department of Respiratory Medicine, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, 261031, Shandong, P. R. China
| | - Baolong Zhou
- School of Pharmacy, Weifang Medical University, Weifang, 261053, Shandong, P. R. China.
| |
Collapse
|
28
|
Li Y, Yang L, Wang Y, Deng Z, Xu S, Xie H, Zhang Y, Li J. Exploring metformin as a candidate drug for rosacea through network pharmacology and experimental validation. Pharmacol Res 2021; 174:105971. [PMID: 34763093 DOI: 10.1016/j.phrs.2021.105971] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/28/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022]
Abstract
Rosacea is a common chronic inflammatory disease that affects the middle of the face. Due to the unclear pathogenesis, the effective treatment options for rosacea remain limited. In this study, weighted gene co-expression network analyses (WGCNA) identified three rosacea-related hub modules, which were involved in immune-, metabolic- and development- related signaling pathways. Next, the key genes from green and brown modules were submitted to CMap database for drug prediction and metformin was identified as a candidate drug for rosacea. Moreover, network pharmacology analysis identified pharmacological targets of metformin and demonstrated that metformin could help in treating rosacea partly by modulating inflammatory and angiogenesis signaling pathways. Finally, we verified the therapeutic role and mechanism of metformin on rosacea in vivo and vitro. We found that metformin treatment significantly improved rosacea-like skin lesions including immune cells infiltration, cytokines/chemokines expression and angiogenesis. Moreover, metformin suppressed LL37- and TNF-α-induced the ROS production and MAPK-NF-κB signal activation in keratinocytes cells. In conclusion, our findings identified and verified metformin as a novel therapeutic candidate for rosacea, and it alleviates the pathological symptoms, possibly by suppressing inflammatory responses, angiogenesis in rosacea.
Collapse
Affiliation(s)
- Yangfan Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Li Yang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Yaling Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - San Xu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Yiya Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; Department of Dermatology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
29
|
Bai H, Yang F, Jiang W, Hu A, Chang H, Zhang Y, Jiang L, Lin S, Lu Z, Zhang C, Cao H. Molybdenum and cadmium co-induce mitophagy and mitochondrial dysfunction via ROS-mediated PINK1/Parkin pathway in Hepa1-6 cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 224:112618. [PMID: 34392151 DOI: 10.1016/j.ecoenv.2021.112618] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 06/13/2023]
Abstract
Excessive molybdenum (Mo) and Cadmium (Cd) can adversely affect health status. However, the correlation between mitophagy and mitochondrial dysfunction caused by Mo and Cd and the underlying mechanisms are still unknown. The aim of this study was to investigate the relationship between mitophagy and mitochondrial dysfunction via the ROS-mediated PINK1/Parkin pathway caused by Mo and Cd. Here, Hepa1-6 cells were incubated with (NH4)6Mo7O24.4 H2O (600.0 μM Mo), CdCl2 (10.0 μM Cd), and the combination of reactive oxygen species (ROS) scavenger (N-acetyl-L-cysteine, NAC, 100.0 μM), or mitophagy inhibitor (Cyclosporin A, CsA, 1.0 μM) for 24 h. Results revealed that Mo or/and Cd elevated the level of intracellular ROS and malondialdehyde (MDA) content, reduced superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px) activities. Additionally, Mo or/and Cd could observably increase the percentage of cells with low membrane potential and decrease the content of ATP, elevate the number of autophagosomes and LC3 puncta, upregulate the mRNA and protein levels of LC3II/LC3I, Parkin, Pink1, VDAC1, downregulate mRNA and protein levels of P62. Moreover, treatments with NAC could significantly alleviate the changes of the above factors co-induced by Mo and Cd, and CsA intensify the changes of the above factors. In summary, our results reveal that Mo and Cd co-exposure can cause oxidative stress and mitophagy via the ROS-mediated PINK1/Parkin pathway in Hepa1-6 cells, and inhibition of mitophagy aggravates Mo and Cd co-induced mitochondrial dysfunction.
Collapse
Affiliation(s)
- He Bai
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China; Medical Research Center, Mudanjiang Medical University, No. 3 Tongxiang street, Aimin District, Mudanjiang 157011, Heilongjiang, PR China
| | - Fan Yang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Wenjuan Jiang
- Application and Extension Department of Animal Husbandry and Fishery Technology of Jiangxi Agricultural Technology Extension Center, No.2, East 2nd Road, Courtyard of Nanchang Municipal Government, East Lake District, Nanchang 330000, Jiangxi, PR China
| | - Aiming Hu
- Ji'an Animal Husbandry and Veterinary Bureau, No.4 Luzhou West Road, Jizhou District, Ji'an 343000, Jiangxi, PR China
| | - Huifeng Chang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Yiling Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Lu Jiang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Shixuan Lin
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Zengting Lu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Huabin Cao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China.
| |
Collapse
|