1
|
Teh HX, Phang SJ, Looi ML, Kuppusamy UR, Arumugam B. Molecular pathways of NF-ĸB and NLRP3 inflammasome as potential targets in the treatment of inflammation in diabetic wounds: A review. Life Sci 2023; 334:122228. [PMID: 37922981 DOI: 10.1016/j.lfs.2023.122228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/23/2023] [Accepted: 10/29/2023] [Indexed: 11/07/2023]
Abstract
Diabetic wounds are slow healing wounds characterized by disordered healing processes and frequently take longer than three months to heal. One of the defining characteristics of impaired diabetic wound healing is an abnormal and unresolved inflammatory response, which is primarily brought on by abnormal macrophage innate immune signaling activation. The persistent inflammatory state in a diabetic wound may be attributed to inflammatory pathways such as nuclear factor kappa B (NF-ĸB) and nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, which have long been associated with inflammatory diseases. Despite the available treatments for diabetic foot ulcers (DFUs) that include debridement, growth factor therapy, and topical anti-bacterial agents, successful wound healing is still hampered. Further understanding of the molecular mechanism of these pathways could be useful in designing potential therapeutic targets for diabetic wound healing. This review provides an update and novel insights into the roles of NF-ĸB and NLRP3 pathways in the molecular mechanism of diabetic wound inflammation and their potential as therapeutic targets in diabetic wound healing.
Collapse
Affiliation(s)
- Huey Xhin Teh
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Shou Jin Phang
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Mee Lee Looi
- Centre for Future Learning, Taylor's University Lakeside Campus, 47500 Subang Jaya, Selangor, Malaysia
| | - Umah Rani Kuppusamy
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Bavani Arumugam
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
2
|
Cazzola M, Rogliani P, Ora J, Calzetta L, Matera MG. Cardiovascular diseases or type 2 diabetes mellitus and chronic airway diseases: mutual pharmacological interferences. Ther Adv Chronic Dis 2023; 14:20406223231171556. [PMID: 37284143 PMCID: PMC10240559 DOI: 10.1177/20406223231171556] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/06/2023] [Indexed: 06/08/2023] Open
Abstract
Chronic airway diseases (CAD), mainly asthma and chronic obstructive pulmonary disease (COPD), are frequently associated with different comorbidities. Among them, cardiovascular disease (CVD) and type 2 diabetes mellitus (T2DM) pose problems for the simultaneous treatment of CAD and comorbidity. Indeed, there is evidence that some drugs used to treat CAD negatively affect comorbidity, and, conversely, some drugs used to treat comorbidity may aggravate CAD. However, there is also growing evidence of some beneficial effects of CAD drugs on comorbidities and, conversely, of the ability of some of those used to treat comorbidity to reduce the severity of lung disease. In this narrative review, we first describe the potential cardiovascular risks and benefits for patients using drugs to treat CAD and the potential lung risks and benefits for patients using drugs to treat CVD. Then, we illustrate the possible negative and positive effects on T2DM of drugs used to treat CAD and the potential negative and positive impact on CAD of drugs used to treat T2DM. The frequency with which CAD and CVD or T2DM are associated requires not only considering the effect that drugs used for one disease condition may have on the other but also providing an opportunity to develop therapies that simultaneously favorably impact both diseases.
Collapse
Affiliation(s)
- Mario Cazzola
- Chair of Respiratory Medicine, Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Paola Rogliani
- Chair of Respiratory Medicine, Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
- Division of Respiratory Medicine, University Hospital Tor Vergata, Rome, Italy
| | - Josuel Ora
- Division of Respiratory Medicine, University Hospital Tor Vergata, Rome, Italy
| | - Luigino Calzetta
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Maria Gabriella Matera
- Chair of Pharmacology, Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
3
|
Gholami M, Klashami ZN, Ebrahimi P, Mahboobipour AA, Farid AS, Vahidi A, Zoughi M, Asadi M, Amoli MM. Metformin and long non-coding RNAs in breast cancer. J Transl Med 2023; 21:155. [PMID: 36849958 PMCID: PMC9969691 DOI: 10.1186/s12967-023-03909-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/21/2023] [Indexed: 03/01/2023] Open
Abstract
Breast cancer (BC) is the second most common cancer and cause of death in women. In recent years many studies investigated the association of long non-coding RNAs (lncRNAs), as novel genetic factors, on BC risk, survival, clinical and pathological features. Recent studies also investigated the roles of metformin treatment as the firstline treatment for type 2 diabetes (T2D) played in lncRNAs expression/regulation or BC incidence, outcome, mortality and survival, separately. This comprehensive study aimed to review lncRNAs associated with BC features and identify metformin-regulated lncRNAs and their mechanisms of action on BC or other types of cancers. Finally, metformin affects BC by regulating five BC-associated lncRNAs including GAS5, HOTAIR, MALAT1, and H19, by several molecular mechanisms have been described in this review. In addition, metformin action on other types of cancers by regulating ten lncRNAs including AC006160.1, Loc100506691, lncRNA-AF085935, SNHG7, HULC, UCA1, H19, MALAT1, AFAP1-AS1, AC026904.1 is described.
Collapse
Affiliation(s)
- Morteza Gholami
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeynab Nickhah Klashami
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Pirooz Ebrahimi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata, Italy
| | | | - Amir Salehi Farid
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Aida Vahidi
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Marziyeh Zoughi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojgan Asadi
- Metabolomics and Genomics Research Center Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa M Amoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Abstract
BACKGROUND Metformin has good anti-hyperglycemic effectiveness, but does not induce hypoglycemia,is very safe, and has become the preferred drug for the treatment of type 2 diabetes. Recently, the other effects of metformin, such as being anti-inflammatory and delaying aging, have also attracted increased attention. METHODS AND RESULTS The relevant literatures on pubmed and other websites for reading, classification and sorting, and did not involve any animal experiments. CONCLUSION Metformin has anti-inflammatory effects through multiple routes, which provides potential therapeutic targets for certain inflammatory diseases, such as neuroinflammation and rheumatoid arthritis. In addition, inflammation is a key component of tumor occurrence and development ; thus, targeted inflammatory intervention is a significant benefit for both cancer prevention and treatment. Therefore, metformin may have further potential for inflammation-related disease prevention and treatmen. However, the inflammatory mechanism is complex; various molecules are connected and influence each other. For example, metformin significantly inhibits p65 nuclear translocation, but pretreatment with compound C, an AMPK inhibitor, abolishes this effect, and silencing of HMGB1 inhibits NF-κB activation . SIRT1 deacetylates FoxO, increasing its transcriptional activity . mTOR in dendritic cells regulates FoxO1 via AKT. The interactions among various molecules should be further explored to clarify their specific mechanisms and provide more direction for the treatment of inflammatory diseases, as well as cancer.
Collapse
|
5
|
Metformin use is not associated with reduced risk of older onset inflammatory bowel disease: a Danish nationwide population-based study. J Gastroenterol 2022; 57:761-769. [PMID: 35780256 DOI: 10.1007/s00535-022-01896-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/13/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Metformin has pleiotropic effects including anti-inflammatory properties and effects on the gut microbiome. It is primarily used in the older population, where the occurrence of inflammatory bowel disease (IBD) is increasing. The aim of this study was to examine whether metformin protects against development of IBD. METHODS In the setting of a Danish nationwide population-based cohort, we conducted a nested case-control study using a new-user active comparator design. For each patient with IBD, we selected 10 IBD-free individuals matched on age, sex, and duration of follow-up. Conditional logistic regression was used to estimate odds ratios (ORs) of IBD. Adjustment included educational level, other immune-mediated inflammatory diseases, and use of dipeptidyl peptidase (DPP)-4 inhibitors and statins. RESULTS Among 302,863 IBD-free new users of oral glucose-lowering drugs, we identified 1271 patients who developed IBD and 12,676 matched IBD-free individuals. Mean age at IBD diagnosis was 66 (SD, 11) years. We found no association between ever use of metformin and risk of IBD, Crohn's disease or ulcerative colitis, adjusted OR 0.95 (95% CI 0.78-1.15), 0.87 (95% CI 0.60-1.26), and 1.04 (95% CI 0.83-1.31), respectively. Neither was the cumulative dose of metformin or the treatment duration with metformin associated with risk of IBD. CONCLUSIONS In this population-based study, we report that despite anti-inflammatory effects and a notable impact on the gut microbiome, metformin use is not associated with reduced risk of older onset IBD.
Collapse
|
6
|
Chen S, Ruan G, Zeng M, Chen T, Cao P, Zhang Y, Li J, Wang X, Li S, Tang S, Lu S, Fan T, Li Y, Han W, Tan J, Ding C, Zhu Z. Association between Metformin Use and Risk of Total Knee Arthroplasty and Degree of Knee Pain in Knee Osteoarthritis Patients with Diabetes and/or Obesity: A Retrospective Study. J Clin Med 2022; 11:4796. [PMID: 36013035 PMCID: PMC9409735 DOI: 10.3390/jcm11164796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/03/2022] Open
Abstract
Objectives: We aimed to examine whether metformin (MET) use is associated with a reduced risk of total knee arthroplasty (TKA) and low severity of knee pain in patients with knee osteoarthritis (OA) and diabetes and/or obesity. Methods: Participants diagnosed with knee OA and diabetes and/or obesity from June 2000 to July 2019 were selected from the information system of a local hospital. Regular MET users were defined as those with recorded prescriptions of MET or self-reported regular MET use for at least 6 months. TKA information was extracted from patients’ surgical records. Knee pain was assessed using the numeric rating scale. Log-binomial regression, linear regression, and propensity score weighting (PSW) were performed for statistical analyses. Results: A total of 862 participants were included in the analyses. After excluding missing data, there were 346 MET non-users and 362 MET users. MET use was significantly associated with a reduced risk of TKA (prevalence ratio: 0.26, 95% CI: 0.15 to 0.45, p < 0.001), after adjustment for age, gender, body mass index, various analgesics, and insurance status. MET use was significantly associated with a reduced degree of knee pain after being adjusted for the above covariates (β: −0.48, 95% CI: −0.91 to −0.05, p = 0.029). There was a significantly accumulative effect of MET use on the reduced risk of TKA. Conclusion: MET can be a potential therapeutic option for OA. Further clinical trials are needed to determine if MET can reduce the risk of TKA and the severity of knee pain in metabolic-associated OA patients.
Collapse
Affiliation(s)
- Shibo Chen
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Guangfeng Ruan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Clinical Research Centre, Guangzhou First People’s Hospital, Guangzhou 510180, China
| | - Muhui Zeng
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Tianyu Chen
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Peihua Cao
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yan Zhang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jia Li
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoshuai Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shengfa Li
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Su’an Tang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shilong Lu
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Tianxiang Fan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yang Li
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Weiyu Han
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jianye Tan
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Zhaohua Zhu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
7
|
Cory TJ, Emmons RS, Yarbro JR, Davis KL, Pence BD. Metformin Suppresses Monocyte Immunometabolic Activation by SARS-CoV-2 Spike Protein Subunit 1. Front Immunol 2021; 12:733921. [PMID: 34858397 PMCID: PMC8631967 DOI: 10.3389/fimmu.2021.733921] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/26/2021] [Indexed: 12/14/2022] Open
Abstract
A hallmark of COVID-19 is a hyperinflammatory state associated with severity. Monocytes undergo metabolic reprogramming and produce inflammatory cytokines when stimulated with SARS-CoV-2. We hypothesized that binding by the viral spike protein mediates this effect, and that drugs which regulate immunometabolism could inhibit the inflammatory response. Monocytes stimulated with recombinant SARS-CoV-2 spike protein subunit 1 showed a dose-dependent increase in glycolytic metabolism associated with production of pro-inflammatory cytokines. This response was dependent on hypoxia-inducible factor-1α, as chetomin inhibited glycolysis and cytokine production. Inhibition of glycolytic metabolism by 2-deoxyglucose (2-DG) or glucose deprivation also inhibited the glycolytic response, and 2-DG strongly suppressed cytokine production. Glucose-deprived monocytes rescued cytokine production by upregulating oxidative phosphorylation, an effect which was not present in 2-DG-treated monocytes due to the known effect of 2-DG on suppressing mitochondrial metabolism. Finally, pre-treatment of monocytes with metformin strongly suppressed spike protein-mediated cytokine production and metabolic reprogramming. Likewise, metformin pre-treatment blocked cytokine induction by SARS-CoV-2 strain WA1/2020 in direct infection experiments. In summary, the SARS-CoV-2 spike protein induces a pro-inflammatory immunometabolic response in monocytes that can be suppressed by metformin, and metformin likewise suppresses inflammatory responses to live SARS-CoV-2. This has potential implications for the treatment of hyperinflammation during COVID-19.
Collapse
Affiliation(s)
- Theodore J. Cory
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Russell S. Emmons
- College of Health Sciences, University of Memphis, Memphis, TN, United States
| | - Johnathan R. Yarbro
- College of Health Sciences, University of Memphis, Memphis, TN, United States
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Kierstin L. Davis
- College of Health Sciences, University of Memphis, Memphis, TN, United States
| | - Brandt D. Pence
- College of Health Sciences, University of Memphis, Memphis, TN, United States
- Center for Nutraceutical and Dietary Supplement Research, University of Memphis, Memphis, TN, United States
| |
Collapse
|
8
|
Ahmadi A, Panahi Y, Johnston TP, Sahebkar A. Antidiabetic drugs and oxidized low-density lipoprotein: A review of anti-atherosclerotic mechanisms. Pharmacol Res 2021; 172:105819. [PMID: 34400317 DOI: 10.1016/j.phrs.2021.105819] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is one of the leading causes of mortality globally. Atherosclerosis is an important step towards different types of cardiovascular disease. The role of oxidized low-density lipoprotein (oxLDL) in the initiation and progression of atherosclerosis has been thoroughly investigated in recent years. Moreover, clinical trials have established that diabetic patients are at a greater risk of developing atherosclerotic plaques. Hence, we aimed to review the clinical and experimental impacts of various classes of antidiabetic drugs on the circulating levels of oxLDL. Metformin, pioglitazone, and dipeptidyl peptidase-4 inhibitors were clinically associated with a suppressive effect on oxLDL in patients with impaired glucose tolerance. However, there is an insufficient number of studies that have clinically evaluated the relationship between oxLDL and newer agents such as agonists of glucagon-like peptide 1 receptor or inhibitors of sodium-glucose transport protein 2. Next, we attempted to explore the multitude of mechanisms that antidiabetic agents exert to counter the undesirable effects of oxLDL in macrophages, endothelial cells, and vascular smooth muscle cells. In general, antidiabetic drugs decrease the uptake of oxLDL by vascular cells and reduce subsequent inflammatory signaling, which prevents macrophage adhesion and infiltration. Moreover, these agents suppress the oxLDL-induced transformation of macrophages into foam cells by either inhibiting oxLDL entrance, or by facilitating its efflux. Thus, the anti-inflammatory, anti-oxidant, and anti-apoptotic properties of antidiabetic agents abrogate changes induced by oxLDL, which can be extremely beneficial in controlling atherosclerosis in diabetic patients.
Collapse
Affiliation(s)
- Ali Ahmadi
- Pharmacotherapy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Yunes Panahi
- Pharmacotherapy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Asutralia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948567, Iran.
| |
Collapse
|
9
|
Metformin Actions on the Liver: Protection Mechanisms Emerging in Hepatocytes and Immune Cells against NASH-Related HCC. Int J Mol Sci 2021; 22:ijms22095016. [PMID: 34065108 PMCID: PMC8126028 DOI: 10.3390/ijms22095016] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is strongly linked to the global epidemic of obesity and type 2 diabetes mellitus (T2DM). Notably, NAFLD can progress from the mildest form of simple steatosis to nonalcoholic steatohepatitis (NASH) that increases the risk for hepatocellular carcinoma (HCC), which is a malignancy with a dismal prognosis and rising incidence in the United States and other developed counties, possibly due to the epidemic of NAFLD. Metformin, the first-line drug for T2DM, has been suggested to reduce risks for several types of cancers including HCC and protect against NASH-related HCC, as revealed by epidemical studies on humans and preclinical studies on animal models. This review focuses on the pathogenesis of NASH-related HCC and the mechanisms by which metformin inhibits the initiation and progression of NASH-related HCC. Since the functional role of immune cells in liver homeostasis and pathogenesis is increasingly appreciated in developing anti-cancer therapies on liver malignancies, we discuss both the traditional targets of metformin in hepatocytes and the recently defined effects of metformin on immune cells.
Collapse
|
10
|
Mbara KC, Mofo Mato PE, Driver C, Nzuza S, Mkhombo NT, Gcwensa SK, Mcobothi EN, Owira PM. Metformin turns 62 in pharmacotherapy: Emergence of non-glycaemic effects and potential novel therapeutic applications. Eur J Pharmacol 2021; 898:173934. [PMID: 33609563 DOI: 10.1016/j.ejphar.2021.173934] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/24/2021] [Accepted: 02/04/2021] [Indexed: 02/07/2023]
Abstract
Metformin is the most commonly prescribed oral antidiabetic medication. Direct/indirect activation of Adenosine Monophosphate-activated protein kinase (AMPK) and non-AMPK pathways, amongst others, are deemed to explain the molecular mechanisms of action of metformin. Metformin is an established insulin receptor sensitising antihyperglycemic agent, is highly affordable, and has superior safety and efficacy profiles. Emerging experimental and clinical evidence suggests that metformin has pleiotropic non-glycemic effects. Metformin appears to have weight stabilising, renoprotective, neuroprotective, cardio-vascular protective, and antineoplastic effects and mitigates polycystic ovarian syndrome. Anti-inflammatory and antioxidant effects of metformin seem to qualify it as an adjunct therapy in treating infectious diseases such as tuberculosis, viral hepatitis, and the current novel Covid-19 infections. So far, metformin is the only prescription medicine relevant to the emerging field of senotherapeutics. Non-glycemic effects of metformin favourable to its repurposing in therapeutic use are hereby discussed.
Collapse
Affiliation(s)
- Kingsley C Mbara
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Pascale E Mofo Mato
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Christine Driver
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Sanelisiwe Nzuza
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Ntokozo T Mkhombo
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Senamile Kp Gcwensa
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Esethu N Mcobothi
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa
| | - Peter Mo Owira
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, University of Kwazulu-Natal, P.O. Box X5401, Durban, South Africa.
| |
Collapse
|
11
|
Crilly NP, Ayeh SK, Karakousis PC. The New Frontier of Host-Directed Therapies for Mycobacterium avium Complex. Front Immunol 2021; 11:623119. [PMID: 33552087 PMCID: PMC7862709 DOI: 10.3389/fimmu.2020.623119] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/14/2020] [Indexed: 01/03/2023] Open
Abstract
Mycobacterium avium complex (MAC) is an increasingly important cause of morbidity and mortality, and is responsible for pulmonary infection in patients with underlying lung disease and disseminated disease in patients with AIDS. MAC has evolved various virulence strategies to subvert immune responses and persist in the infected host. Current treatment for MAC is challenging, requiring a combination of multiple antibiotics given over a long time period (for at least 12 months after negative sputum culture conversion). Moreover, even after eradication of infection, many patients are left with residual lung dysfunction. In order to address similar challenges facing the management of patients with tuberculosis, recent attention has focused on the development of novel adjunctive, host-directed therapies (HDTs), with the goal of accelerating the clearance of mycobacteria by immune defenses and reducing or reversing mycobacterial-induced lung damage. In this review, we will summarize the evidence supporting specific adjunctive, HDTs for MAC, with a focus on the repurposing of existing immune-modulatory agents targeting a variety of different cellular pathways. We also highlight areas meriting further investigation.
Collapse
Affiliation(s)
- Nathan P Crilly
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Samuel K Ayeh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Petros C Karakousis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
12
|
Role of metformin in various pathologies: state-of-the-art microcapsules for improving its pharmacokinetics. Ther Deliv 2020; 11:733-753. [PMID: 32967584 DOI: 10.4155/tde-2020-0102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Metformin was originally derived from a botanical ancestry and became the most prescribed, first-line therapy for Type 2 diabetes in most countries. In the last century, metformin was discovered twice for its antiglycemic properties in addition to its antimalarial and anti-influenza effects. Metformin exhibits flip-flop pharmacokinetics with limited oral bioavailability. This review outlines metformin pharmacokinetics, pharmacodynamics and recent advances in polymeric particulate delivery systems as a potential tool to target metformin delivery to specific tissues/organs. This interesting biguanide is being rediscovered this century for multiple clinical indications as anticancer, anti-aging, anti-inflammatory, anti-Alzheimer's and much more. Microparticulate delivery systems of metformin may improve its oral bioavailability and optimize the therapeutic goals expected.
Collapse
|
13
|
Systemic Oxidative Stress and Visceral Adipose Tissue Mediators of NLRP3 Inflammasome and Autophagy Are Reduced in Obese Type 2 Diabetic Patients Treated with Metformin. Antioxidants (Basel) 2020; 9:antiox9090892. [PMID: 32967076 PMCID: PMC7555880 DOI: 10.3390/antiox9090892] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/17/2022] Open
Abstract
Obesity is a low-grade inflammatory condition affecting a range of individuals, from metabolically healthy obese (MHO) subjects to type 2 diabetes (T2D) patients. Metformin has been shown to display anti-inflammatory properties, though the underlying molecular mechanisms are unclear. To study whether the effects of metformin are mediated by changes in the inflammasome complex and autophagy in visceral adipose tissue (VAT) of obese patients, a biopsy of VAT was obtained from a total of 68 obese patients undergoing gastric bypass surgery. The patients were clustered into two groups: MHO patients and T2D patients treated with metformin. Patients treated with metformin showed decreased levels of all analyzed serum pro-inflammatory markers (TNFα, IL6, IL1β and MCP1) and a downwards trend in IL18 levels associated with a lower production of oxidative stress markers in leukocytes (mitochondrial ROS and myeloperoxidase (MPO)). A reduction in protein levels of MCP1, NFκB, NLRP3, ASC, ATG5, Beclin1 and CHOP and an increase in p62 were also observed in the VAT of the diabetic group. This downregulation of both the NLRP3 inflammasome and autophagy in VAT may be associated with the improved inflammatory profile and leukocyte homeostasis seen in obese T2D patients treated with metformin with respect to MHO subjects and endorses the cardiometabolic protective effect of this drug.
Collapse
|
14
|
Thangaraju P, Varthya SB, Venkatesan S. Target/therapies for chronic recurrent erythema nodosum leprosum. Indian J Pharmacol 2020; 52:222-226. [PMID: 32874007 PMCID: PMC7446680 DOI: 10.4103/ijp.ijp_788_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/12/2020] [Accepted: 03/25/2020] [Indexed: 12/04/2022] Open
Abstract
A Type 2 lepra reaction or erythema nodosum leprosum is an anticipated complication in the lepromatous spectrum of leprosy cases. It is an example of an immune complex-mediated complement activated disease (Type III hypersensitivity reaction). Hence, we tried to target the inflammatory mediators and the mental stressors for the possible management strategies.
Collapse
Affiliation(s)
- Pugazhenthan Thangaraju
- Department of Pharmacology, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| | - Shoban Babu Varthya
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Sajitha Venkatesan
- Department of Microbiology, All India Institute of Medical Sciences, Raipur, Chhattisgarh, India
| |
Collapse
|
15
|
He Y, Ren E, Lu Z, Chen H, Qin Z, Wang J, He M, Liu G, Zheng L, Zhao J. Rational engineering of ferritin nanocages for targeted therapy of osteoarthritis. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 28:102210. [DOI: 10.1016/j.nano.2020.102210] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 02/08/2020] [Accepted: 04/10/2020] [Indexed: 12/26/2022]
|
16
|
The comparision of glybenclamide and metformin-loaded bacterial cellulose/gelatin nanofibres produced by a portable electrohydrodynamic gun for diabetic wound healing. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.109844] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
17
|
Naicker N, Sigal A, Naidoo K. Metformin as Host-Directed Therapy for TB Treatment: Scoping Review. Front Microbiol 2020; 11:435. [PMID: 32411100 PMCID: PMC7201016 DOI: 10.3389/fmicb.2020.00435] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/02/2020] [Indexed: 11/30/2022] Open
Abstract
Tuberculosis (TB) disease is an international health concern caused by the bacteria Mycobacterium tuberculosis (Mtb). Evolution of multi-drug-resistant strains may cause bacterial persistence, rendering existing antibiotics ineffective. Hence, development of new or repurposing of currently approved drugs to fight Mtb in combination with existing antibiotics is urgently needed to cure TB which is refractory to current therapy. The shortening of TB therapy and reduction in lung injury can be achieved using adjunctive host-directed therapies. There is a wide range of probable candidates which include numerous agents permitted for the treatment of other diseases. One potential candidate is metformin, a Food and Drug Administration (FDA)-approved drug used to treat type 2 diabetes mellitus (DM). However, there is a scarcity of evidence supporting the biological basis for the effect of metformin as a host-directed therapy for TB. This scoping review summarizes the current body of evidence and outlines scientific gaps that need to be addressed in determining the potential role of metformin as a host-directed therapy.
Collapse
Affiliation(s)
- Nikita Naicker
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Alex Sigal
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Kogieleum Naidoo
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- MRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
18
|
Marcucci F, Romeo E, Caserta CA, Rumio C, Lefoulon F. Context-Dependent Pharmacological Effects of Metformin on the Immune System. Trends Pharmacol Sci 2020; 41:162-171. [DOI: 10.1016/j.tips.2020.01.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/14/2019] [Accepted: 01/06/2020] [Indexed: 12/20/2022]
|
19
|
Karadeniz Z, Aynacıoğlu AŞ, Bilir A, Tuna MY. Inhibition of midkine by metformin can contribute to its anticancer effects in malignancies: A proposal mechanism of action of metformin in context of endometrial cancer prevention and therapy. Med Hypotheses 2019; 134:109420. [PMID: 31634770 DOI: 10.1016/j.mehy.2019.109420] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/20/2019] [Accepted: 09/30/2019] [Indexed: 01/10/2023]
Abstract
Metformin, a drug widely used in the treatment of type II diabetes mellitus (T2DM), has been the focus of interest as a potential therapeutic agent for certain types of malignancies, including gynaecological cancers [i.e. endometrial cancer (EC)]. Although the exact mechanism behind the potential anticancer activity of metformin is still not completely understood, certain studies have suggested that different effects on cell functions, such as inhibition of cell migration, apoptosis and tumor cell proliferation, are involved in its preventive and therapeutic effects in certain types of malignancies, including EC. In contrast, midkine (MK), a heparin-binding growth factor and cytokine, which induces carcinogenesis and chemoresistance, promotes the development and progression of many malignant tumours by increasing diverse cell functions such as cell proliferation, cell survival and antiapoptotic activities via mainly the activation of phosphatidyl inositol 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways. The same pathways are also subject to certain therapeutic effects of metformin, although this cytokine and this drug have some different mechanism of action pathways as well. Taken together, MK and metformin appear to have opposite effects in various biological processes such as apoptosis, cell proliferation, cell survival, cell migration, and angiogenesis. On the other hand, MK activates PI3K and MAPK cell signal pathways, whereas metformin inhibits these two pathways. It seems likely that almost all the pathways and cell functions, which play important roles in malignancies, are inhibited by metformin and activated by MK. Given the opposite relationship between the actions of metformin and MK, we hypothesize that metformin may act like a novel MK inhibitor in some malignancies. We also discuss the possible relationship between metformin and MK in the context of EC, the most common gynecological cancer worldwide, which incidence is rising rapidly, in parallel with the increase in obesity, T2DM and insulin resistance. In this respect, the therapeutic use of metformin may improve the survival of EC or other cancers, via inhibiting or overcoming the unwanted effects of MK in carcinogenesis.
Collapse
Affiliation(s)
- Zeliha Karadeniz
- Department of Gynecology and Obstetrics, Istanbul Aydin University, Medical Faculty, Florya Main Campus, Kücükcekmece, 34295 Istanbul, Turkey
| | - A Şükrü Aynacıoğlu
- Department of Medical Pharmacology, Istanbul Aydin University, Medical Faculty, Florya Main Campus, Kücükcekmece, 34295 Istanbul, Turkey.
| | - Ayhan Bilir
- Department of Histology and Embryology, Istanbul Aydin University, Medical Faculty, Florya Main Campus, Kücükcekmece, 34295 Istanbul, Turkey
| | - M Yakup Tuna
- Department of Anatomy, Istanbul Aydin University, Medical Faculty, Florya Main Campus, Kücükcekmece, 34295 Istanbul, Turkey
| |
Collapse
|
20
|
Foretz M, Guigas B, Viollet B. Understanding the glucoregulatory mechanisms of metformin in type 2 diabetes mellitus. Nat Rev Endocrinol 2019; 15:569-589. [PMID: 31439934 DOI: 10.1038/s41574-019-0242-2] [Citation(s) in RCA: 396] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2019] [Indexed: 02/07/2023]
Abstract
Despite its position as the first-line drug for treatment of type 2 diabetes mellitus, the mechanisms underlying the plasma glucose level-lowering effects of metformin (1,1-dimethylbiguanide) still remain incompletely understood. Metformin is thought to exert its primary antidiabetic action through the suppression of hepatic glucose production. In addition, the discovery that metformin inhibits the mitochondrial respiratory chain complex 1 has placed energy metabolism and activation of AMP-activated protein kinase (AMPK) at the centre of its proposed mechanism of action. However, the role of AMPK has been challenged and might only account for indirect changes in hepatic insulin sensitivity. Various mechanisms involving alterations in cellular energy charge, AMP-mediated inhibition of adenylate cyclase or fructose-1,6-bisphosphatase 1 and modulation of the cellular redox state through direct inhibition of mitochondrial glycerol-3-phosphate dehydrogenase have been proposed for the acute inhibition of gluconeogenesis by metformin. Emerging evidence suggests that metformin could improve obesity-induced meta-inflammation via direct and indirect effects on tissue-resident immune cells in metabolic organs (that is, adipose tissue, the gastrointestinal tract and the liver). Furthermore, the gastrointestinal tract also has a major role in metformin action through modulation of glucose-lowering hormone glucagon-like peptide 1 and the intestinal bile acid pool and alterations in gut microbiota composition.
Collapse
Affiliation(s)
- Marc Foretz
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Centre, Leiden, Netherlands
| | - Benoit Viollet
- INSERM, U1016, Institut Cochin, Paris, France.
- CNRS, UMR8104, Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
21
|
Ma X, Jiang Z, Wang Z, Zhang Z. Administration of metformin alleviates atherosclerosis by promoting H2S production via regulating CSE expression. J Cell Physiol 2019; 235:2102-2112. [PMID: 31338841 DOI: 10.1002/jcp.29112] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/20/2019] [Indexed: 12/31/2022]
Abstract
The therapeutic effect of metformin (Met) on atherosclerosis was studied here. Effects of methionine and Met on the induction of inflammatory response and H2 S expression in peritoneal macrophages were evaluated. Enzyme-linked immunosorbent assay, immunohistochemistry assay, western blot, and quantitative reverse transcription polymerase chain reaction were conducted to observe the levels of cystathionine γ-lyase (CSE), DNA methyltransferases 1 (DNMT1), DNMT3a, DNMT3b, tumor necrosis factor (TNF- α), interleukin 1b (IL-1β), and hydrogen sulfide (H 2 S). Luciferase and bisulfite sequencing assays were also utilized to evaluate the CSE promoter activity as well as the methylation status of CSE in transfected cells. Methionine significantly elevated Hcy, TNF-a, H 2 S, and IL-1β expression while decreasing the level of CSE in C57BL/6 mice. In contrary, co-treatment with Methionine and Met reduced the detrimental effect of Methionine. Homocysteine (Hcy) decreased H 2 S expression while promoting the synthesis of IL-1β and TNF-α in THP-1 and raw264.7 cells. Treatment of THP-1 and raw264.7 cells with methionine and Met reduced the activity of methionine in dose dependently. Moreover, Hcy increased the expression of DNMT and elevated the level of methylation in the CSE promoter, whereas the co-treatment with methionine and Met attenuated the effects of Hcy. Methionine significantly decreased plasma level of CSE while increasing the severity of inflammatory responses and plasma level of Hcy, which in turn suppressed H 2 S synthesis and enhanced DNA hypermethylation of CSE promoter to promote the pathogenesis of atherosclerosis. In contrary, co-treatment with methionine and Met reduced the detrimental effect of methionine.
Collapse
Affiliation(s)
- Xiaofeng Ma
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan, University of South China, Hengyang, Hunan, China.,Department of Cardiology, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan, China
| | - Zhisheng Jiang
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan, University of South China, Hengyang, Hunan, China
| | - Zuo Wang
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan, University of South China, Hengyang, Hunan, China
| | - Zhuhua Zhang
- Department of Cardiology, Beijing Hospital of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
22
|
He X, Wang L, Chen XF, Liang Q, Wang WQ, Lin AQ, Yi L, Wang Y, Gao Q. Metformin improved oxidized low-density lipoprotein-impaired mitochondrial function and increased glucose uptake involving Akt-AS160 pathway in raw264.7 macrophages. Chin Med J (Engl) 2019; 132:1713-1722. [PMID: 31268904 PMCID: PMC6759109 DOI: 10.1097/cm9.0000000000000333] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Macrophage accumulation in the vascular wall is a hallmark of atherosclerosis. Studies showed that shifting of oxidized lipids-induced inflammatory macrophages towards an anti-inflammatory phenotype by promoting oxidative metabolism attenuated atherosclerosis progression. Therefore, this study aimed to investigate whether metformin, which has ameliorated atherosclerosis in animal models and clinical trials, modulated oxidized low-density lipoprotein (Ox-LDL) induced inflammatory status in macrophages by regulating cellular oxidative metabolism. METHODS Murine raw264.7 macrophages were incubated with Ox-LDL (50 μg/mL) in the presence or absence of metformin (15 μmol/L) for 24 h. Real-time polymerase chain reaction was used to quantify the transcription of classically activated (M1) pro-inflammatory and alternatively activated (M2) anti-inflammatory markers and mitochondrial DNA copy numbers. Cellular reactive oxygen species (ROS) production and mitochondrial membrane potential were detected by immunofluorescence. Cellular adenosine triphosphate (ATP) synthesis, glucose uptake, and lactic acid production were measured by commercial kit and normalized to cellular lysates. Western blotting analysis was performed to detect the expression of mitochondrial fusion/fission related proteins, enzymes mediating lipid metabolism and signaling pathway of glucose transport. Differences between groups were analyzed using one-way analysis of variance. RESULTS Metformin improved Ox-LDL-impaired anti-inflammatory phenotype in raw264.7 macrophages as shown by up-regulated transcription of anti-inflammatory markers including interleukin 10 (0.76 ± 0.04 vs. 0.94 ± 0.01, P = 0.003) and Resistin-like molecule alpha (0.67 ± 0.08 vs. 1.78 ± 0.34, P = 0.030). Conversely, Ox-LDL-diminished phosphorylation of Akt was up-regulated by metformin treatment (0.47 ± 0.05 vs. 1.02 ± 0.08, P = 0.040), associated with an improvement of mitochondrial function, characterized by decreased ROS generation (2.50 ± 0.07 vs. 2.15 ± 0.04, P = 0.040), increased lipid oxidation, and elevated cellular ATP production (0.026 ± 0.001 vs. 0.035 ± 0.003, P = 0.020). Moreover, metformin-mediated Akt activation increased Akt substrate of 160 kDa (AS160) phosphorylation (0.51 ± 0.04 vs. 1.03 ± 0.03, P = 0.0041), promoted membrane translocation of glucose transporter 1, and increased glucose influx into the cells (4.78 ± 0.04 vs. 5.47 ± 0.01, P < 0.001). CONCLUSION This study suggested that targeting macrophage metabolism with new or existing drugs had therapeutic potential for the prevention and treatment of diabetes-accelerated atherosclerosis.
Collapse
Affiliation(s)
- Xuan He
- Medical School of Nanjing University, Nanjing, Jiangsu 210093, China
| | - Lei Wang
- Medical School of Nanjing University, Nanjing, Jiangsu 210093, China
| | - Xiu-Fang Chen
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Qiao Liang
- Medical School of Nanjing University, Nanjing, Jiangsu 210093, China
| | - Wen-Qing Wang
- Medical School of Nanjing University, Nanjing, Jiangsu 210093, China
| | - An-Qi Lin
- Medical School of Nanjing University, Nanjing, Jiangsu 210093, China
| | - Long Yi
- Medical School of Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yong Wang
- Medical School of Nanjing University, Nanjing, Jiangsu 210093, China
| | - Qian Gao
- Medical School of Nanjing University, Nanjing, Jiangsu 210093, China
| |
Collapse
|
23
|
Nguyen TT, Ung TT, Li S, Lian S, Xia Y, Park SY, Do Jung Y. Metformin inhibits lithocholic acid-induced interleukin 8 upregulation in colorectal cancer cells by suppressing ROS production and NF-kB activity. Sci Rep 2019; 9:2003. [PMID: 30765814 PMCID: PMC6376015 DOI: 10.1038/s41598-019-38778-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 10/22/2018] [Indexed: 12/29/2022] Open
Abstract
Metformin, an inexpensive, well-tolerated oral agent that is a commonly used first-line treatment for type 2 diabetes, has become the focus of intense research as a potential anticancer agent. In this study, we describe the inhibitory effect of metformin in interleukin 8 (IL-8) upregulation by lithocholic acid (LCA) in HCT116 colorectal cancer (CRC) cells. Pharmacological inhibition studies indicated that reactive oxygen species (ROS) were involved in LCA-induced IL-8 upregulation through activation of the transcription factor NF-κB. Metformin was demonstrated to block LCA-stimulated ROS production, in turn suppressing NF-κB signaling that was critical for IL-8 upregulation. An NADPH oxidase assay proved that the inhibitory effect of metformin on ROS production was derived from its strong suppression of NADPH oxidase, a key producer of ROS in cells. Compared with conditioned media (CM) derived from HCT116 cells treated with LCA, CM derived from HCT116 cells pretreated with metformin and then treated with LCA lost all stimulatory effect on endothelial cell proliferation and tubelike formation. In conclusion, metformin inhibited NADPH oxidase, which in turn suppressed ROS production and NF-κB activation to prevent IL-8 upregulation stimulated by LCA; this prevention thus obstructed endothelial cell proliferation and tubelike formation.
Collapse
Affiliation(s)
- Thi Thinh Nguyen
- Department of Biochemistry, Chonnam National University Medical School, Seoyang Ro 264, Hwasun, Jeonnam, 58138, Korea
| | - Trong Thuan Ung
- Department of Biochemistry, Chonnam National University Medical School, Seoyang Ro 264, Hwasun, Jeonnam, 58138, Korea
| | - Shinan Li
- Department of Biochemistry, Chonnam National University Medical School, Seoyang Ro 264, Hwasun, Jeonnam, 58138, Korea
| | - Sen Lian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yong Xia
- Department of Urology, New York University School of Medicine, New York, NY, 10010, USA
| | - Sun Young Park
- Department of Biochemistry, Chonnam National University Medical School, Seoyang Ro 264, Hwasun, Jeonnam, 58138, Korea
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Seoyang Ro 264, Hwasun, Jeonnam, 58138, Korea.
| |
Collapse
|
24
|
Li J, Shen X. Effect of rosiglitazone on inflammatory cytokines and oxidative stress after intensive insulin therapy in patients with newly diagnosed type 2 diabetes. Diabetol Metab Syndr 2019; 11:35. [PMID: 31073335 PMCID: PMC6499940 DOI: 10.1186/s13098-019-0432-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/26/2019] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To evaluate the effect of insulin sensitizer on inflammatory cytokines and oxidative stress in patients with newly diagnosed type 2 diabetes mellitus (T2DM). METHODS After intensive insulin therapy, patients with newly diagnosed T2DM were continuously treated with either insulin sensitizer or insulin for 48 weeks, and then their inflammatory cytokine and oxidative stress levels were measured. RESULTS Tumor necrosis factor alpha (TNF-α), interleukin (IL)-6, hypersensitive C reactive protein (hs-CRP), malondialdehyde (MDA), and 8-iso-prostaglandin F2α (8-iso-PGF2α) levels of the rosiglitazone (RSG) group and the rosiglitazone combined with metformin (RSG + metformin) group were significantly reduced after the treatments (P < 0.05). Hs-CRP, MDA, and 8-iso-PGF2α levels of the metformin group were significantly reduced after the treatments (P < 0.05). Superoxide dismutase (SOD) and total antioxidant capacity (TAC) were significantly increased after the treatments in all three groups (P < 0.05 and P < 0.01). CONCLUSION Early application of insulin sensitizers improved inflammation and oxidative stress in patients with newly diagnosed T2DM.
Collapse
Affiliation(s)
- Juan Li
- Department of Emergency, Zhongshan Hospital Xiamen University, Xiamen, 361004 Fujian China
| | - Xingping Shen
- Department of Endocrinology, Zhongshan Hospital Xiamen University, Xiamen, 361004 Fujian China
| |
Collapse
|
25
|
Chen YC, Kuo CH, Tsai YM, Lin YC, Hsiao HP, Chen BH, Chen YT, Wang SL, Hung CH. Suppressive effects of metformin on T-helper 1-related chemokines expression in the human monocytic leukemia cell line THP-1. Endocr Res 2018; 43:228-234. [PMID: 29630425 DOI: 10.1080/07435800.2018.1460605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF THE STUDY Type 1 and type 2 diabetes mellitus (DM) are chronic T-cell-mediated inflammatory diseases. Metformin is a widely used drug for type 2 DM that reduces the need for insulin in type 1 DM. However, whether metformin has an anti-inflammatory effect for treating DM is unknown. We investigated the anti-inflammatory mechanism of metformin in the human monocytic leukemia cell line THP-1. MATERIALS AND METHODS The human monocytic leukemia cell line THP-1 was pretreated with metformin and stimulated with lipopolysaccharide (LPS). The production of T-helper (Th)-1-related chemokines including interferon-γ-induced protein-10 (IP-10) and monocyte chemoattractant protein-1 (MCP-1), Th2-related chemokine macrophage-derived chemokine, and the proinflammatory chemokine tumor necrosis factor-α was measured using enzyme-linked immunosorbent assay. Intracellular signaling pathways were investigated using Western blot analysis and chromatin immunoprecipitation assay. RESULTS Metformin suppressed LPS-induced IP-10 and MCP-1 production as well as LPS-induced phosphorylation of c-Jun N-terminal kinase (JNK), p38, extracellular signal-regulated kinase (ERK), and nuclear factor-kappa B (NF-κB). Moreover, metformin suppressed LPS-induced acetylation of histones H3 and H4 at the IP-10 promoter. CONCLUSIONS Metformin suppressed the production of Th1-related chemokines IP-10 and MCP-1 in THP-1 cells. Suppressive effects of metformin on IP-10 production might be attributed at least partially to the JNK, p38, ERK, and NF-κB pathways as well as to epigenetic regulation through the acetylation of histones H3 and H4. These results indicated the therapeutic anti-inflammatory potential of metformin.
Collapse
Affiliation(s)
- Yen-Chun Chen
- a Department of Pediatrics , Kaohsiung Municipal Hsiao-Kang Hospital , Kaohsiung , Taiwan
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Chang-Hung Kuo
- c Ta-Kuo Clinic , Kaohsiung , Taiwan
- d Department of Pediatrics , Kaohsiung Municipal Ta-Tung Hospital , Kaohsiung , Taiwan
| | - Ying-Ming Tsai
- e Department of Internal Medicine , Kaohsiung Municipal Ta-Tung Hospital , Kaohsiung , Taiwan
- f Graduate Institute of Clinical Medicine, College of Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Yi-Ching Lin
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
- f Graduate Institute of Clinical Medicine, College of Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan
- g Department of Laboratory Medicine, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Hui-Pin Hsiao
- a Department of Pediatrics , Kaohsiung Municipal Hsiao-Kang Hospital , Kaohsiung , Taiwan
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
- h Department of Pediatrics, Faculty of Medicine, College of Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Bai-Hsiun Chen
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
- g Department of Laboratory Medicine, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
- h Department of Pediatrics, Faculty of Medicine, College of Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Yi-Ting Chen
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Shih-Ling Wang
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Chih-Hsing Hung
- a Department of Pediatrics , Kaohsiung Municipal Hsiao-Kang Hospital , Kaohsiung , Taiwan
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
- h Department of Pediatrics, Faculty of Medicine, College of Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan
- i Research Center for Environmental Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan
| |
Collapse
|
26
|
Stek C, Allwood B, Walker NF, Wilkinson RJ, Lynen L, Meintjes G. The Immune Mechanisms of Lung Parenchymal Damage in Tuberculosis and the Role of Host-Directed Therapy. Front Microbiol 2018; 9:2603. [PMID: 30425706 PMCID: PMC6218626 DOI: 10.3389/fmicb.2018.02603] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/11/2018] [Indexed: 12/20/2022] Open
Abstract
Impaired lung function is common in people with a history of tuberculosis. Host-directed therapy added to tuberculosis treatment may reduce lung damage and result in improved lung function. An understanding of the pathogenesis of pulmonary damage in TB is fundamental to successfully predicting which interventions could be beneficial. In this review, we describe the different features of TB immunopathology that lead to impaired lung function, namely cavities, bronchiectasis, and fibrosis. We discuss the immunological processes that cause lung damage, focusing on studies performed in humans, and using chest radiograph abnormalities as a marker for pulmonary damage. We highlight the roles of matrix metalloproteinases, neutrophils, eicosanoids and cytokines, like tumor necrosis factor-α and interleukin 1β, as well as the role of HIV co-infection. Finally, we focus on various existing drugs that affect one or more of the immunological mediators of lung damage and could therefore play a role as host-directed therapy.
Collapse
Affiliation(s)
- Cari Stek
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium.,Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Brian Allwood
- Division of Pulmonology, Department of Medicine, Stellenbosch University, Stellenbosch, South Africa
| | - Naomi F Walker
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Robert J Wilkinson
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Medicine, University of Cape Town, Cape Town, South Africa.,Department of Medicine, Imperial College London, London, United Kingdom.,Francis Crick Institute, London, United Kingdom
| | - Lutgarde Lynen
- Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Graeme Meintjes
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
27
|
Kewcharoenwong C, Prabowo SA, Bancroft GJ, Fletcher HA, Lertmemongkolchai G. Glibenclamide Reduces Primary Human Monocyte Functions Against Tuberculosis Infection by Enhancing M2 Polarization. Front Immunol 2018; 9:2109. [PMID: 30283449 PMCID: PMC6157405 DOI: 10.3389/fimmu.2018.02109] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/28/2018] [Indexed: 12/31/2022] Open
Abstract
Tuberculosis (TB) is a global public health problem, which is caused by Mycobacterium tuberculosis (Mtb). Type 2 diabetes mellitus (T2DM) is one of the leading predisposing factors for development of TB after HIV/AIDS. Glibenclamide is a widely used anti-diabetic drug in low and middle-income countries where the incidence of TB is very high. In a human macrophage cell line, glibenclamide, a K+ATP-channel blocker, promoted alternative activation of macrophages by enhancing expression of the M2 marker CD206 during M2 polarization. M2 macrophages are considered poorly microbicidal and associated with TB susceptibility. Here, we investigated the effect of glibenclamide on M1 and M2 phenotypes of primary human monocytes and further determined whether specific drug treatment for T2DM individuals influences the antibacterial function of monocytes in response to mycobacterial infection. We found that glibenclamide significantly reduced M1 (HLA-DR+ and CD86+) surface markers and TNF-α production on primary human monocytes against mycobacterial infection. In contrast, M2 (CD163+ and CD206+) surface markers and IL-10 production were enhanced by pretreatment with glibenclamide. Additionally, reduction of bactericidal activity also occurred when primary human monocytes from T2DM individuals who were being treated with glibenclamide were infected with Mtb in vitro, consistent with the cytokine responses. We conclude that glibenclamide reduces M1 and promotes M2 polarization leading to impaired bactericidal ability of primary human monocytes of T2DM individuals in response to Mtb and may lead to increased susceptibility of T2DM individuals to TB and other bacterial infectious diseases.
Collapse
Affiliation(s)
- Chidchamai Kewcharoenwong
- Mekong Health Science Research Institute, Khon Kaen, Thailand.,Faculty of Associated Medical Sciences, The Centre for Research and Development of Medical Diagnostic Laboratories, Khon Kaen University, Khon Kaen, Thailand
| | - Satria A Prabowo
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Gregory J Bancroft
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Helen A Fletcher
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Tuberculosis Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ganjana Lertmemongkolchai
- Mekong Health Science Research Institute, Khon Kaen, Thailand.,Faculty of Associated Medical Sciences, The Centre for Research and Development of Medical Diagnostic Laboratories, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
28
|
Bulatova N, Kasabri V, Qotineh A, Al-Athami T, Yousef AM, AbuRuz S, Momani M, Zayed A. Effect of metformin combined with lifestyle modification versus lifestyle modification alone on proinflammatory-oxidative status in drug-naïve pre-diabetic and diabetic patients: A randomized controlled study. Diabetes Metab Syndr 2018; 12:257-267. [PMID: 29221717 DOI: 10.1016/j.dsx.2017.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 11/22/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Targeting biomarkers of oxidative-proinflammatory stress may result in improvement of modifiable metabolic syndrome, pre-diabetes and diabetes risk factors and subsequent risk reduction. METHODS 64 newly diagnosed antihyperglycemic treatment-naïve prediabetic and type 2 diabetes mellitus (T2DM) patients were randomly assigned using block design to either metformin combined with therapeutic lifestyle changes (TLC) or TLC alone. Body mass index (BMI), waist circumference, blood pressure, fasting plasma glucose (FPG), glycated hemoglobin (HbA1c), fasting lipid profile, plasma oxidative status and tumor necrosis factor (TNF)-α were measured at baseline, after 3 months and after 6 months from baseline. RESULTS Except for HbA1c, baseline values did not differ significantly between the two groups. The post 3-months relative reductions in BMI (P=0.014) and HbA1c (P=0.037) in metformin combined with TLC intervention were significantly greater than those in TLC alone group. TNFα plasma levels were decreased significantly vs. baseline by metformin combined with TLC intervention (-22.90±46.76%, P=0.01). Conversely, TLC alone basically worsened proinflammatory status (42.40±40.82 %), P<0.001. Metformin with TLC treatment effected a therapeutic decrement of the oxidative stress (-15.44±35.32%, P=0.029 vs. baseline) unlike TLC alone (61.49±122.66%, P=0.01 vs. baseline). Both interventions' effects were sustained in the 6-month follow up periods. CONCLUSION In both intervention groups, the relative changes in plasma TNFα were significantly correlated (P<0.01) with systolic blood pressure and the relative changes in oxidative stress were markedly correlated (P<0.05) with total cholesterol.
Collapse
Affiliation(s)
- Nailya Bulatova
- School of Pharmacy, The University of Jordan, Queen Rania Street, Amman 11942, Jordan
| | - Violet Kasabri
- School of Pharmacy, The University of Jordan, Queen Rania Street, Amman 11942, Jordan.
| | - Amenah Qotineh
- School of Pharmacy, The University of Jordan, Queen Rania Street, Amman 11942, Jordan
| | - Taiba Al-Athami
- School of Pharmacy, The University of Jordan, Queen Rania Street, Amman 11942, Jordan
| | - Al-Motassem Yousef
- School of Pharmacy, The University of Jordan, Queen Rania Street, Amman 11942, Jordan
| | - Salah AbuRuz
- School of Pharmacy, The University of Jordan, Queen Rania Street, Amman 11942, Jordan; College of Pharmacy, Al Ain University of Science and Technology, AL Ain, Abu Dhabi, United Arab Emirates
| | - Munther Momani
- School of Medicine, The University of Jordan, Queen Rania Street, Amman 11942, Jordan
| | - Aymen Zayed
- School of Medicine, The University of Jordan, Queen Rania Street, Amman 11942, Jordan
| |
Collapse
|
29
|
Witkowski M, Tabaraie T, Steffens D, Friebel J, Dörner A, Skurk C, Witkowski M, Stratmann B, Tschoepe D, Landmesser U, Rauch U. MicroRNA-19a contributes to the epigenetic regulation of tissue factor in diabetes. Cardiovasc Diabetol 2018; 17:34. [PMID: 29477147 PMCID: PMC6389222 DOI: 10.1186/s12933-018-0678-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/17/2018] [Indexed: 12/21/2022] Open
Abstract
Background Diabetes mellitus is characterized by chronic vascular disorder and presents a main risk factor for cardiovascular mortality. In particular, hyperglycaemia and inflammatory cytokines induce vascular circulating tissue factor (TF) that promotes pro-thrombotic conditions in diabetes. It has recently become evident that alterations of the post-transcriptional regulation of TF via specific microRNA(miR)s, such as miR-126, contribute to the pathogenesis of diabetes and its complications. The endothelial miR-19a is involved in vascular homeostasis and atheroprotection. However, its role in diabetes-related thrombogenicity is unknown. Understanding miR-networks regulating procoagulability in diabetes may help to develop new treatment options preventing vascular complications. Methods and results Plasma of 44 patients with known diabetes was assessed for the expression of miR-19a, TF protein, TF activity, and markers for vascular inflammation. High miR-19a expression was associated with reduced TF protein, TF-mediated procoagulability, and vascular inflammation based on expression of vascular adhesion molecule-1 and leukocyte count. We found plasma expression of miR-19a to strongly correlate with miR-126. miR-19a reduced the TF expression on mRNA and protein level in human microvascular endothelial cells (HMEC) as well as TF activity in human monocytes (THP-1), while anti-miR-19a increased the TF expression. Interestingly, miR-19a induced VCAM expression in HMEC. However, miR-19a and miR-126 co-transfection reduced total endothelial VCAM expression and exhibited additive inhibition of a luciferase reporter construct containing the F3 3′UTR. Conclusions While both miRs have differential functions on endothelial VCAM expression, miR-19a and miR-126 cooperate to exhibit anti-thrombotic properties via regulating vascular TF expression. Modulating the post-transcriptional control of TF in diabetes may provide a future anti-thrombotic and anti-inflammatory therapy. Electronic supplementary material The online version of this article (10.1186/s12933-018-0678-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marco Witkowski
- Charité Centrum 11, Depart. of Cardiology, Campus Benjamin Franklin, Charité University Medicine Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Termeh Tabaraie
- Charité Centrum 11, Depart. of Cardiology, Campus Benjamin Franklin, Charité University Medicine Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Daniel Steffens
- Charité Centrum 11, Depart. of Cardiology, Campus Benjamin Franklin, Charité University Medicine Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Julian Friebel
- Charité Centrum 11, Depart. of Cardiology, Campus Benjamin Franklin, Charité University Medicine Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Andrea Dörner
- Charité Centrum 11, Depart. of Cardiology, Campus Benjamin Franklin, Charité University Medicine Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Carsten Skurk
- Charité Centrum 11, Depart. of Cardiology, Campus Benjamin Franklin, Charité University Medicine Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Mario Witkowski
- Institute of Microbiology and Infection Immunology, Charité University Medicine Berlin, Berlin, Germany
| | - Bernd Stratmann
- Heart and Diabetes Center NRW, Ruhr University of Bochum, Bad Oeynhausen, Germany
| | - Diethelm Tschoepe
- Heart and Diabetes Center NRW, Ruhr University of Bochum, Bad Oeynhausen, Germany
| | - Ulf Landmesser
- Charité Centrum 11, Depart. of Cardiology, Campus Benjamin Franklin, Charité University Medicine Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Ursula Rauch
- Charité Centrum 11, Depart. of Cardiology, Campus Benjamin Franklin, Charité University Medicine Berlin, Hindenburgdamm 30, 12200, Berlin, Germany.
| |
Collapse
|
30
|
Ziegelhoeffer T, Heil M, Fischer S, Fernández B, Schaper W, Preissner KT, Deindl E, Pagel JI. Role of early growth response 1 in arteriogenesis: Impact on vascular cell proliferation and leukocyte recruitment in vivo. Thromb Haemost 2017; 107:562-74. [DOI: 10.1160/th11-07-0490] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 12/13/2011] [Indexed: 02/07/2023]
Abstract
SummaryBased on previous findings that early growth response 1 (Egr-1) participates in leukocyte recruitment and cell proliferation in vitro, this study was designed to investigate its mode of action during arteriogenesis in vivo. In a model of peripheral arteriogenesis, Egr-1 was significantly upregulated in growing collaterals of wild-type (WT) mice, both on mRNA and protein level. Egr-1−/− mice demonstrated delayed arteriogenesis after femoral artery ligation. They further showed increased levels of monocytes and granulocytes in the circulation, but reduced levels in adductor muscles under baseline conditions. After femoral artery ligation, elevated numbers of macrophages were detected in the perivascular zone of collaterals in Egr-1−/− mice and mRNA of leukocyte recruitment mediators was upregulated. Other Egr family members (Egr-2 to -4) were significantly upregulated only in Egr-1−/− mice, suggesting a mechanism of counterbalancing Egr-1 deficiency. Moreover, splicing factor-1, downregulated in WT mice after femoral artery ligation in the process of increased vascular cell proliferation, was upregulated in Egr-1−/− mice. αSM-actin on the other hand, significantly downregulated in WT mice, showed no differential expression in Egr-1−/− mice. While cell cycle regulator cyclin E and cdc20 were upregulated in Egr-1−/− mice, cyclin D1 expression decreased below the detection limit in collaterals, and the proliferation marker ki67 was not differentially expressed. In conclusion, compensation for deficiency in Egr-1 function in leukocyte recruitment can presumably be mediated by other transcription factors; however, Egr-1 is indispensable for effective vascular cell cycle progression in arteriogenesis.
Collapse
|
31
|
Li C, Xue Y, Xi YR, Xie K. Progress in the application and mechanism of metformin in treating non-small cell lung cancer. Oncol Lett 2017; 13:2873-2880. [PMID: 28529553 PMCID: PMC5431600 DOI: 10.3892/ol.2017.5862] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 01/04/2017] [Indexed: 12/17/2022] Open
Abstract
At present, the incidence and mortality of lung cancer demonstrate an increasing trend. Non-small cell lung cancer (NSCLC) accounts for ~80–85% of all lung cancer cases. Therefore, developing novel and more effective treatments is of great importance. The use of combination therapies, where several anticancer agents are used together, is a promising strategy. Recent studies demonstrate that metformin, which has been utilized for treating diabetes mellitus for >50 years, has antitumor effects in numerous types of cancer including NSCLC. Its antitumor effects can be direct and indirect, and it is able to synergize with other physical therapies including targeted anticancer therapy, chemotherapy and radiotherapy. The present review discusses how metformin affects cellular energy metabolism in NSCLC, the mechanism of its antitumor action and its synergy with other therapies. Information and analysis are provided in the present review to stimulate further studies on metformin as an adjunct anticancer treatment.
Collapse
Affiliation(s)
- Chan Li
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Yang Xue
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Yu-Rong Xi
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Ke Xie
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
32
|
Chen CZ, Hsu CH, Li CY, Hsiue TR. Insulin use increases risk of asthma but metformin use reduces the risk in patients with diabetes in a Taiwanese population cohort. J Asthma 2017; 54:1019-1025. [PMID: 28135899 DOI: 10.1080/02770903.2017.1283698] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Recent reports have suggested that insulin promotes airway smooth muscle contraction and enhances airway hyperresponsiveness, which are cardinal features of asthma. In contrast, metformin can reduce both airway inflammatory and remodeling properties. However, these results are all from in vitro and animal studies. This study investigated whether diabetes and various antidiabetic agents associate with the risk of asthma. METHODS We used a retrospective population-based cohort study using Taiwan's National Health Insurance claim database from 2000 to 2010 and a Cox proportional hazards regression model to compare the incidence of asthma between patients with diabetes (n = 19,428) and a matched non-diabetic group (n = 38,856). We also used a case-control study nested from the above cohort including 1,982 incident cases of asthma and 1,982 age- and sex-matched controls. A time density sampling technique was used to assess the effects of various antidiabetic agents on the risk of asthma. RESULTS The incidence of asthma was significantly higher in the diabetic cohort than that in the non-diabetic cohort after adjustment for age, sex, and obesity, with a hazard ratio of 1.30 (95% confidence interval [CI]: 1.24-1.38). Insulin was found to increase the risk of asthma among diabetic patients (odds ratio [OR] 2.23; 95% CI: 1.52-3.58). In contrast, the use of metformin correlated with a decreased risk of asthma (OR 0.75; 95% CI: 0.60-0.95). CONCLUSIONS Individuals with diabetes are at an increased risk of asthma. Insulin may further increase the risk of asthma, but the risk could possibly be reduced by using metformin.
Collapse
Affiliation(s)
- Chiung-Zuei Chen
- a Division of Chest Medicine, Department of Internal Medicine , National Cheng Kung University Medical College and Hospital , Tainan , Taiwan
| | - Chih-Hui Hsu
- a Division of Chest Medicine, Department of Internal Medicine , National Cheng Kung University Medical College and Hospital , Tainan , Taiwan
| | - Chung-Yi Li
- b Institute of Public Health, National Cheng Kung University Medical College , Tainan , Taiwan
| | - Tzuen-Ren Hsiue
- a Division of Chest Medicine, Department of Internal Medicine , National Cheng Kung University Medical College and Hospital , Tainan , Taiwan
| |
Collapse
|
33
|
Lei Y, Yi Y, Liu Y, Liu X, Keller ET, Qian CN, Zhang J, Lu Y. Metformin targets multiple signaling pathways in cancer. CHINESE JOURNAL OF CANCER 2017; 36:17. [PMID: 28126011 PMCID: PMC5270304 DOI: 10.1186/s40880-017-0184-9] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/21/2016] [Indexed: 12/20/2022]
Abstract
Metformin, an inexpensive and well-tolerated oral agent commonly used in the first-line treatment of type 2 diabetes, has become the focus of intense research as a candidate anticancer agent. Here, we discuss the potential of metformin in cancer therapeutics, particularly its functions in multiple signaling pathways, including AMP-activated protein kinase, mammalian target of rapamycin, insulin-like growth factor, c-Jun N-terminal kinase/mitogen-activated protein kinase (p38 MAPK), human epidermal growth factor receptor-2, and nuclear factor kappaB pathways. In addition, cutting-edge targeting of cancer stem cells by metformin is summarized.
Collapse
Affiliation(s)
- Yong Lei
- Key Laboratory of Longevity and Ageing-related Diseases, Ministry of Education, Nanning, 530021, Guangxi, P. R. China.,Center for Translational Medicine, Guangxi Medical University, 14th Floor, Pharmacology and Biomedical Sciences Building, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, P. R. China
| | - Yanhua Yi
- School for International Education, Guangxi Medical University, Nanning, 530021, Guangxi, P. R. China
| | - Yang Liu
- Key Laboratory of Longevity and Ageing-related Diseases, Ministry of Education, Nanning, 530021, Guangxi, P. R. China.,Center for Translational Medicine, Guangxi Medical University, 14th Floor, Pharmacology and Biomedical Sciences Building, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, P. R. China
| | - Xia Liu
- Key Laboratory of Longevity and Ageing-related Diseases, Ministry of Education, Nanning, 530021, Guangxi, P. R. China.,Center for Translational Medicine, Guangxi Medical University, 14th Floor, Pharmacology and Biomedical Sciences Building, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, P. R. China
| | - Evan T Keller
- Department of Urology and Pathology, School of Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chao-Nan Qian
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Jian Zhang
- Key Laboratory of Longevity and Ageing-related Diseases, Ministry of Education, Nanning, 530021, Guangxi, P. R. China. .,Center for Translational Medicine, Guangxi Medical University, 14th Floor, Pharmacology and Biomedical Sciences Building, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, P. R. China. .,Department of Urology and Pathology, School of Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Yi Lu
- Key Laboratory of Longevity and Ageing-related Diseases, Ministry of Education, Nanning, 530021, Guangxi, P. R. China. .,Center for Translational Medicine, Guangxi Medical University, 14th Floor, Pharmacology and Biomedical Sciences Building, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, P. R. China.
| |
Collapse
|
34
|
Kassem AA, Issa DAE, Kotry GS, Farid RM. Thiolated alginate-based multiple layer mucoadhesive films of metformin forintra-pocket local delivery: in vitro characterization and clinical assessment. Drug Dev Ind Pharm 2016; 43:120-131. [PMID: 27589817 DOI: 10.1080/03639045.2016.1224895] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Periodontal disease broadly defines group of conditions in which the supportive structure of the tooth (periodontium) is destroyed. Recent studies suggested that the anti-diabetic drug metformin hydrochloride (MF) has an osteogenic effect and is beneficial for the management of periodontitis. OBJECTIVE Development of strong mucoadhesive multiple layer film loading small dose of MF for intra-pocket application. METHODOLOGY Multiple layer film was developed by double casting followed by compression method. Either 6% carboxy methyl cellulose sodium (CMC) or sodium alginate (ALG) constituted the inner drug (0.6%) loaded layer. Thiolated sodium alginate (TSA; 2 or 4%) constituted the outer drug free layers to enhance mucoadhesion and achieve controlled drug release. Optimized formulation was assessed clinically on 20 subjects. RESULTS Films were uniform, thin and hard enough for easy insertion into periodontal pockets. Based on water uptake and in vitro drug release, CMC based film with 4% TSA as an outer layer was the optimized formulation with enhanced mucoadhesion and controlled drug release (83.73% over 12 h). SEM showed the effective fabrication of the triple layer film in which connective lines between the layers could be observed. FTIR examination suggests possibility of hydrogen bonding between the -NH groups of metformin and -OH groups of CMC. DSC revealed the presence of MF mainly in the amorphous form. Clinical results indicated improvement of all clinical parameters six months post treatment. CONCLUSION The results suggested that local application of the mucoadhesive multiple layer films loaded with metformin hydrochloride was able to manage moderate chronic periodontitis.
Collapse
Affiliation(s)
- Abeer Ahmed Kassem
- a Department of Pharmaceutical Sciences, Faculty of Pharmacy , Princess Nourah bint Abdulrahman University , Riyadh , Saudi Arabia.,b Department of Pharmaceutics, Faculty of Pharmacy , Alexandria University , Alexandria , Egypt
| | - Doaa Ahmed Elsayed Issa
- c Department of Pharmaceutical Sciences, Faculty of Pharmacy , Beirut Arab University , Beirut , Lebanon.,d Department of Pharmaceutical Chemistry, Faculty of Pharmacy , Alexandria University , Alexandria , Egypt
| | - Gehan Sherif Kotry
- e Department of Oral Medicine, Periodontology, Oral Diagnosis and Radiology, Faculty of Dentistry , Alexandria University , Alexandria , Egypt
| | - Ragwa Mohamed Farid
- f Department of Pharmaceutics, Faculty of Pharmacy and Drug Manufacturing , Pharos University in Alexandria , Alexandria , Egypt
| |
Collapse
|
35
|
Sheta A, Elsakkar M, Hamza M, Solaiman A. Effect of metformin and sitagliptin on doxorubicin-induced cardiotoxicity in adult male albino rats. Hum Exp Toxicol 2016; 35:1227-1239. [PMID: 26818447 DOI: 10.1177/0960327115627685] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The use of doxorubicin (DOX) as an antitumor therapeutic agent is limited due to its cardiotoxic effects. Metformin (Met) and sitagliptin (Sitg) are suggested to improve cardiac function. The present study aimed to determine the potential protective effects of Met and Sitg on DOX-induced cardiotoxicity. Rats were divided into six groups: groups I, II, and III received normal saline, Met, and Sitg, respectively. Groups IV, V, and VI received DOX only, Met + DOX, and Sitg + DOX, respectively. Heart tissue was used for biochemical assays which measured cardiac reduced glutathione (GSH), thiobarbituric acid reactive substances (TBARS), and tumor necrosis factor α (TNF-α). Serum creatinine kinase (CK) and lactate dehydrogenase (LDH) were also measured. The heart apex was prepared for histological (hematoxylin and eosin) and immunohistochemical examination. Intoxication of DOX was associated with a significant elevation in serum CK-MB and LDH, reduction in cardiac GSH, and increased TBARS and TNF-α compared to the controls. Administration of Met or Sitg to DOX-intoxicated rats suppressed serum CK-MB and LDH. Moreover, cardiac GSH was elevated with decreased TBARS and TNF-α. These results were confirmed by histological study. Met and Sitg caused inhibition of caspase 3 and upregulation of B-cell lymphoma 2 (Bcl-2) expression in DOX-intoxicated animals. Sitg was found to exert a significantly better protective effect compared to that of Met. It was concluded that Sitg might be more effective than Met in reducing myocardial injury in DOX-induced cardiotoxicity in rats.
Collapse
Affiliation(s)
- A Sheta
- 1 Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - M Elsakkar
- 2 Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - M Hamza
- 2 Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - A Solaiman
- 3 Department of Histology and Cell Biology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
36
|
Rockel JS, Kapoor M. Autophagy: controlling cell fate in rheumatic diseases. Nat Rev Rheumatol 2016; 12:517-31. [DOI: 10.1038/nrrheum.2016.92] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
37
|
Salazar JJ, Ennis WJ, Koh TJ. Diabetes medications: Impact on inflammation and wound healing. J Diabetes Complications 2016; 30:746-52. [PMID: 26796432 PMCID: PMC4834268 DOI: 10.1016/j.jdiacomp.2015.12.017] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 12/25/2022]
Abstract
Chronic wounds are a common complication in patients with diabetes that often lead to amputation. These non-healing wounds are described as being stuck in a persistent inflammatory state characterized by accumulation of pro-inflammatory macrophages, cytokines and proteases. Some medications approved for management of type 2 diabetes have demonstrated anti-inflammatory properties independent of their marketed insulinotropic effects and thus have underappreciated potential to promote wound healing. In this review, the potential for insulin, metformin, specific sulfonylureas, thiazolidinediones, and dipeptidyl peptidase-4 inhibitors to promote healing is evaluated by reviewing human and animal studies on inflammation and wound healing. The available evidence indicates that diabetic medications have potential to prevent wounds from becoming arrested in the inflammatory stage of healing and to promote wound healing by downregulating pro-inflammatory cytokines, upregulating growth factors, lowering matrix metalloproteinases, stimulating angiogenesis, and increasing epithelization. However, no clinical recommendations currently exist on the potential for specific diabetic medications to impact healing of chronic wounds. Thus, we encourage further research that may guide physicians on providing personalized diabetes treatments that achieve glycemic goals while promoting healing in patients with chronic wounds.
Collapse
Affiliation(s)
- Jay J Salazar
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - William J Ennis
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, USA; Center for Tissue Repair and Regeneration, University of Illinois at Chicago, Chicago, IL, USA
| | - Timothy J Koh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA; Center for Tissue Repair and Regeneration, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
38
|
Wang H, Li T, Chen S, Gu Y, Ye S. Neutrophil Extracellular Trap Mitochondrial DNA and Its Autoantibody in Systemic Lupus Erythematosus and a Proof-of-Concept Trial of Metformin. Arthritis Rheumatol 2016; 67:3190-200. [PMID: 26245802 DOI: 10.1002/art.39296] [Citation(s) in RCA: 239] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 07/16/2015] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Neutrophil extracellular traps (NETs) have been shown to play an important role in systemic lupus erythematosus (SLE) pathogenesis by activating plasmacytoid dendritic cells (PDCs) and the type I interferon (IFN) pathway. NETs composed of self-DNA are considered to be of nuclear origin and are a major source of anti-DNA autoantibody generation. This study was undertaken to evaluate whether mitochondrial DNA (mtDNA) resides in NETs, to evaluate whether mtDNA and anti-mtDNA antibodies cause dysregulation of the PDC-IFNα pathway, and to investigate the clinical implication in SLE. METHODS Patients with SLE (n = 102), patients with rheumatoid arthritis (n = 30), and healthy donors (n = 40) were enrolled in in vitro studies. NETs were generated from phorbol 12-myristate 13-acetate (PMA)-stimulated peripheral neutrophils. Immunofluorescence staining was used to detect NET formation ex vivo and in lupus nephritis renal biopsy samples. The mtDNA levels and type I IFN-inducible gene scores were measured by quantitative polymerase chain reaction. Anti-mtDNA antibodies, anti-double-stranded DNA (anti-dsDNA) antibodies, and IFNα were detected by enzyme-linked immunosorbent assay. Purified PDCs were stimulated by isolated NETs, mtDNA, or dsDNA, combined with anti-mtDNA or dsDNA IgG, or other culture conditions. Additional patients with SLE (n = 113) were enrolled in a proof-of-concept trial. We evaluated the efficacy and safety of metformin on a background of corticosteroids and conventional immunosuppressive agents in patients with mild or moderate lupus. The primary end point was the efficacy of metformin for reducing disease flare. RESULTS We detected mtDNA in NETs, and anti-mtDNA antibody levels were elevated in SLE patients compared with controls and significantly correlated with IFN scores and the disease activity index. The presence of anti-mtDNA antibodies was disproportionately associated with lupus nephritis, and correlated better than anti-dsDNA antibody levels with the lupus nephritis activity index. Mitochondrial DNA was deposited in NETs in lupus nephritis renal biopsy specimens. In addition, mtDNA/anti-mtDNA were greater inducers of PDC IFNα production via Toll-like receptor engagement than dsDNA/anti-dsDNA. We assessed the effect of metformin on down-regulating the NET mtDNA-PDC-IFNα pathway. Metformin decreased PMA-induced NET formation and CpG-stimulated PDC IFNα generation. A proof-of-concept trial of metformin add-on treatment of mild or moderate SLE resulted in decreases in clinical flares, prednisone exposure, and body weight. CONCLUSION Our findings establish a link between mtDNA in NETs, anti-mtDNA antibodies, and PDC IFNα pathogenesis in SLE, and highlight that specific strategies to down-regulate this pathway, such as treatment with metformin, may be new approaches to treat SLE.
Collapse
Affiliation(s)
- Haiting Wang
- Renji Hospital South Campus and Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ting Li
- Renji Hospital South Campus and Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Sheng Chen
- Renji Hospital South Campus and Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yueying Gu
- Renji Hospital South Campus and Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Shuang Ye
- Renji Hospital South Campus and Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
Metformin Changes the Relationship between Blood Monocyte Toll-Like Receptor 4 Levels and Nonalcoholic Fatty Liver Disease-Ex Vivo Studies. PLoS One 2016; 11:e0150233. [PMID: 26930651 PMCID: PMC4773077 DOI: 10.1371/journal.pone.0150233] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 02/10/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Toll-like receptor 4 (TLR4) contributes to the development of NAFLD (nonalcoholic fatty liver disease) and MetS (metabolic syndrome). It is unclear whether anti-diabetic metformin affects TLR4 expression on blood monocytes, thereby protecting or improving inflammatory parameters. Therefore, we investigated TLR4 in patients with NAFLD meeting different sets of MetS criteria and linked the results with the disease burden. METHODS 70 subjects were characterized and divided into three groups: (I) healthy individuals, (II) nonobese with NAFLD and without MetS, and (III) prediabetic, obese with NAFLD and MetS. We determined the concentrations of IL-1β, IL-6, TNFα, and monocyte TLR4 levels in fresh blood as well as in blood cultures with or without metformin supplementation. RESULTS The characteristics of the study groups revealed a significant association between NAFLD and BMI, MetS and inflammatory parameters, and TLR4. In ex vivo studies, 100 μM of metformin decreased the TLR4 level by 19.9% (II group) or by 35% (III group) as well as IL-1β and TNFα production. A stepwise multiple regression analysis highlighted a strong effect of metformin on attenuation of the link between TLR4 and NAFLD, and TNFα. CONCLUSION We concluded that, by attenuation of the blood monocyte TLR4 level, metformin reduced their inflammatory potential-critical after recruitment these cells into liver. However, this finding should be confirmed after in vivo metformin administration.
Collapse
|
40
|
Rezagholizadeh L, Pourfarjam Y, Nowrouzi A, Nakhjavani M, Meysamie A, Ziamajidi N, Nowrouzi PS. Effect of Cichorium intybus L. on the expression of hepatic NF-κB and IKKβ and serum TNF-α in STZ- and STZ+ niacinamide-induced diabetes in rats. Diabetol Metab Syndr 2016; 8:11. [PMID: 26877773 PMCID: PMC4752748 DOI: 10.1186/s13098-016-0128-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 02/01/2016] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Inflammation is an early event in the development of diabetes type 2 (T2D). Cichorium intybus L. (chicory) possesses anti-inflammatory action. We compared the anti-inflammatory aspect of aqueous chicory seed extract (CSE) in early and late stage T2D in rats. METHODS Wistar albino rats were divided into nine final groups (n = 6). Three main groups consisted of non-diabetic (Control), early stage diabetes (ET2D; niacinamide/streptozotocin, i.e., NIA/STZ), and late stage diabetes (LT2D; STZ). Within each main group, a subgroup was treated with CSE (125 mg/kg; i.p.); within each diabetic group (STZ and NIA/STZ) a subgroup received metformin (100 mg/kg; i.p.); another subgroup in STZ group received aspirin (120 mg/kg; oral). After 21 days, fasting blood glucose (FBS), insulin, and TNF-α level were measured in serum; IKKβ and NF-κB (p65) mRNA and protein expression were evaluated by real time PCR and Western blotting; p65 DNA binding activity was determined by ELISA, in liver tissue. RESULTS The mRNA and protein expression levels of IKKβ, and P65 genes increased in both stages of T2D (p < 0.01); CSE decreased their expression (p < 0.001, mRNAs; p < 0.05, proteins). The increased DNA-binding capacity of NF-κB (p < 0.0001) in diabetes was lowered by CSE (p < 0.001). The effect of CSE was limited to ET2D requiring insulin. CONCLUSIONS The anti-inflammatory action of CSE is due to a direct modulation of cytokine expression. The dependency of chicory action on the presence of insulin indicates its usefulness in the early stages of diabetes and for the purpose of preventing and delaying diabetes onset.
Collapse
Affiliation(s)
- Lotfollah Rezagholizadeh
- />Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Enghelab Avenue, Poursina Street, Tehran, Iran
| | - Yasin Pourfarjam
- />Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Enghelab Avenue, Poursina Street, Tehran, Iran
| | - Azin Nowrouzi
- />Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Enghelab Avenue, Poursina Street, Tehran, Iran
| | - Manuchehr Nakhjavani
- />Endocrinology and Metabolism Research Center, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Alipasha Meysamie
- />Department of Community and Preventive Medicine, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrin Ziamajidi
- />Department of Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Peyman S. Nowrouzi
- />Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Enghelab Avenue, Poursina Street, Tehran, Iran
| |
Collapse
|
41
|
Hyperreactivity of Blood Leukocytes in Patients with NAFLD to Ex Vivo Lipopolysaccharide Treatment Is Modulated by Metformin and Phosphatidylcholine but Not by Alpha Ketoglutarate. PLoS One 2015; 10:e0143851. [PMID: 26629827 PMCID: PMC4667886 DOI: 10.1371/journal.pone.0143851] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 11/09/2015] [Indexed: 01/29/2023] Open
Abstract
INTRODUCTION AND AIMS Toll-like receptor 4 and proinflammatory cytokines play a central role in the progression of nonalcoholic fatty liver disease. We investigated IL-1, IL-6 and TNFα production and toll-like receptor 4 in both--obese and lean patients with non-alcoholic fatty liver disease who met different sets of metabolic syndrome criteria and linked the results with the disease burden. MATERIALS AND METHODS 95 subjects were divided into four groups depending on the following criteria: presence or absence of metabolic syndrome and/or non-alcoholic fatty liver disease, glucose tolerance (prediabetes or normoglycemia) and BMI value (obese or lean). We determined the levels of IL-1β, IL-6, TNFα, and monocyte toll-like receptor 4 expression in fresh blood as well as in blood cultures treated with lipopolysaccharide with or without metformin, alphaketoglutarate or phosphatidylcholine supplementation. RESULTS The blood leukocytes of patients with non-alcoholic fatty liver disease are hypersensitive to lipopolysaccharide treatment and produce elevated levels of pro-inflammatory cytokines in response to ex vivo treatment with lipopolysaccharide. Moreover, they overexpress toll-like receptor-4. Hyperreactivity was typical mainly for obese patients with non-alcoholic fatty liver disease together with metabolic syndrome and decreased with the severity of disease. Metformin was the most effective in attenuation of hyperreactivity in all groups of patients with non-alcoholic fatty liver disease, but in obese patients the effectiveness of metformin was weaker than in lean. The reduction of cytokine level by metformin was accompanied by the decrease in toll-like receptor-4 expression. phosphatidylcholine also attenuated hyperreactivity to lipopolysaccharide but mainly in obese patients. Alpha ketoglutarate did not modulate cytokines' level and toll-like receptor 4 expression in non-alcoholic fatty liver disease patients. CONCLUSIONS Metformin and phosphatidylcholine attenuated lipopolysaccharide induced toll-like receptor 4 overexpression and overproduction of pro-inflammatory cytokines; however, their efficacy depended on combined presence of non-alcoholic fatty liver disease, metabolic syndrome and obesity.
Collapse
|
42
|
Edén D, Siegbahn A, Mokhtari D. Tissue factor/factor VIIa signalling promotes cytokine-induced beta cell death and impairs glucose-stimulated insulin secretion from human pancreatic islets. Diabetologia 2015; 58:2563-72. [PMID: 26271343 PMCID: PMC4589554 DOI: 10.1007/s00125-015-3729-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/20/2015] [Indexed: 01/19/2023]
Abstract
AIMS/HYPOTHESIS Patients diagnosed with type 1 or type 2 diabetes have elevated levels of coagulation factor VIIa (FVIIa) and its receptor tissue factor (TF) in their bloodstream. This may affect the fate of the beta cells. We aimed to study the effects of TF/FVIIa signalling on cytokine-induced beta cell death and islet function in vitro. METHODS Human pancreatic islets and MIN-6 beta cells were used to study TF mRNA and protein expression using real-time PCR, immunoblotting and flow cytometry. The effects of TF/FVIIa on cytokine-induced beta cell death were studied in MIN-6 cells and human pancreatic islets using cell-death ELISA and propidium iodide and cleaved caspase-3 staining. Effects of TF/FVIIa on the phosphorylation of p38, extracellular signal-regulated kinase and c-Jun N-terminal kinase (JNK) were investigated by immunoblotting. Glucose-stimulated insulin secretion (GSIS) from human islets was measured with an insulin ELISA. RESULTS A combination of the cytokines IL-1β, TNF-α and IFN-γ induced TF expression in human pancreatic islets and in beta cells. TF/FVIIa did not affect basal beta cell death but, independently of downstream coagulation activity, augmented beta cell death in response to cytokines. The effect of TF/FVIIa on cytokine-induced beta cell death was found to be dependent on the stress kinase JNK, since FVIIa addition potentiated cytokine-induced JNK activation and JNK inhibition abolished the effect of TF/FVIIa on cytokine-induced beta cell death. Moreover, TF/FVIIa signalling resulted in inhibition of GSIS from human pancreatic islets. CONCLUSIONS/INTERPRETATION These results indicate that TF/FVIIa signalling has a negative effect on beta cell function and promotes beta cell death in response to cytokines.
Collapse
Affiliation(s)
- Desirée Edén
- Department of Medical Sciences, Clinical Chemistry, Science for Life Laboratory, University Hospital, Uppsala University, Entr. 61 3rd floor, S-751 85, Uppsala, Sweden
| | - Agneta Siegbahn
- Department of Medical Sciences, Clinical Chemistry, Science for Life Laboratory, University Hospital, Uppsala University, Entr. 61 3rd floor, S-751 85, Uppsala, Sweden
| | - Dariush Mokhtari
- Department of Medical Sciences, Clinical Chemistry, Science for Life Laboratory, University Hospital, Uppsala University, Entr. 61 3rd floor, S-751 85, Uppsala, Sweden.
| |
Collapse
|
43
|
Kiran D, Podell BK, Chambers M, Basaraba RJ. Host-directed therapy targeting the Mycobacterium tuberculosis granuloma: a review. Semin Immunopathol 2015; 38:167-83. [PMID: 26510950 PMCID: PMC4779125 DOI: 10.1007/s00281-015-0537-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/13/2015] [Indexed: 12/16/2022]
Abstract
Infection by the intracellular bacterial pathogen Mycobacterium tuberculosis (Mtb) is a major cause of morbidity and mortality worldwide. Slow progress has been made in lessening the impact of tuberculosis (TB) on human health, especially in parts of the world where Mtb is endemic. Due to the complexity of TB disease, there is still an urgent need to improve diagnosis, prevention, and treatment strategies to control global spread of disease. Active research targeting avenues to prevent infection or transmission through vaccination, to diagnose asymptomatic carriers of Mtb, and to improve antimicrobial drug treatment responses is ongoing. However, this research is hampered by a relatively poor understanding of the pathogenesis of early infection and the factors that contribute to host susceptibility, protection, and the development of active disease. There is increasing interest in the development of adjunctive therapy that will aid the host in responding to Mtb infection appropriately thereby improving the effectiveness of current and future drug treatments. In this review, we summarize what is known about the host response to Mtb infection in humans and animal models and highlight potential therapeutic targets involved in TB granuloma formation and resolution. Strategies designed to shift the balance of TB granuloma formation toward protective rather than destructive processes are discussed based on our current knowledge. These therapeutic strategies are based on the assumption that granuloma formation, although thought to prevent the spread of the tubercle bacillus within and between individuals contributes to manifestations of active TB disease in human patients when left unchecked. This effect of granuloma formation favors the spread of infection and impairs antimicrobial drug treatment. By gaining a better understanding of the mechanisms by which Mtb infection contributes to irreversible tissue damage, down regulates protective immune responses, and delays tissue healing, new treatment strategies can be rationally designed. Granuloma-targeted therapy is advantageous because it allows for the repurpose of existing drugs used to treat other communicable and non-communicable diseases as adjunctive therapies combined with existing and future anti-TB drugs. Thus, the development of adjunctive, granuloma-targeted therapy, like other host-directed therapies, may benefit from the availability of approved drugs to aid in treatment and prevention of TB. In this review, we have attempted to summarize the results of published studies in the context of new innovative approaches to host-directed therapy that need to be more thoroughly explored in pre-clinical animal studies and in human clinical trials.
Collapse
Affiliation(s)
- Dilara Kiran
- Department of Microbiology, Immunology and Pathology, Metabolism of Infectious Diseases Laboratory and Mycobacteria Research Laboratories, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 200 West Lake Street, 1619 Campus Delivery, Fort Collins, CO, 80523-1619, USA
| | - Brendan K Podell
- Department of Microbiology, Immunology and Pathology, Metabolism of Infectious Diseases Laboratory and Mycobacteria Research Laboratories, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 200 West Lake Street, 1619 Campus Delivery, Fort Collins, CO, 80523-1619, USA
| | - Mark Chambers
- Department of Bacteriology, Animal and Plant Health Agency (APHA), Woodham Lane, New Haw, Addlestone, Surrey, KT15 3NB, UK.,School of Veterinary Medicine Faculty of Health and Medical Sciences, University of Surrey, Vet School Main Building, Daphne Jackson Road, Guildford, GU2 7AL, UK
| | - Randall J Basaraba
- Department of Microbiology, Immunology and Pathology, Metabolism of Infectious Diseases Laboratory and Mycobacteria Research Laboratories, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 200 West Lake Street, 1619 Campus Delivery, Fort Collins, CO, 80523-1619, USA.
| |
Collapse
|
44
|
Characterization of Peripheral Immune Cell Subsets in Patients with Acute and Chronic Cerebrovascular Disease: A Case-Control Study. Int J Mol Sci 2015; 16:25433-49. [PMID: 26512654 PMCID: PMC4632808 DOI: 10.3390/ijms161025433] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 09/29/2015] [Accepted: 10/19/2015] [Indexed: 01/14/2023] Open
Abstract
Immune cells (IC) play a crucial role in murine stroke pathophysiology. However, data are limited on the role of these cells in ischemic stroke in humans. We therefore aimed to characterize and compare peripheral IC subsets in patients with acute ischemic stroke/transient ischemic attack (AIS/TIA), chronic cerebrovascular disease (CCD) and healthy volunteers (HV). We conducted a case-control study of patients with AIS/TIA (n = 116) or CCD (n = 117), and HV (n = 104) who were enrolled at the University Hospital Würzburg from 2010 to 2013. We determined the expression and quantity of IC subsets in the three study groups and performed correlation analyses with demographic and clinical parameters. The quantity of several IC subsets differed between the AIS/TIA, CCD, and HV groups. Several clinical and demographic variables independently predicted the quantity of IC subsets in patients with AIS/TIA. No significant changes in the quantity of IC subsets occurred within the first three days after AIS/TIA. Overall, these findings strengthen the evidence for a pathophysiologic role of IC in human ischemic stroke and the potential use of IC-based biomarkers for the prediction of stroke risk. A comprehensive description of IC kinetics is crucial to enable the design of targeted treatment strategies.
Collapse
|
45
|
Cavaglieri RC, Day RT, Feliers D, Abboud HE. Metformin prevents renal interstitial fibrosis in mice with unilateral ureteral obstruction. Mol Cell Endocrinol 2015; 412:116-22. [PMID: 26067231 DOI: 10.1016/j.mce.2015.06.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/04/2015] [Accepted: 06/04/2015] [Indexed: 01/08/2023]
Abstract
Unilateral ureteral obstruction causes important tubulo-interstitial fibrosis in the kidney. Metformin reduces fibrosis in mice with diabetic nephropathy. We examined the effects of metformin in a mouse model of unilateral ureteral obstruction (UUO). Expression of inflammation and fibrosis markers was studied by immunohistochemistry, immunoblot and quantitative real-time polymerase chain reaction. Seven days after UUO, kidneys presented dilated tubules, expansion of the tubulo-interstitial compartment, and significant infiltration of inflammatory cells. Macrophage infiltration and inflammation markers expression were increased in obstructed kidneys and reduced by metformin. Metformin reduced expression of extracellular matrix proteins and profibrotic factor TGFβ in obstructed kidneys, measured by immunohistochemistry. Interstitial fibroblast activation was evident in obstructed kidneys and ameliorated by metformin. UUO did not affect adenosine monophosphate-activated kinase (AMPK) activity, but metformin activated AMPK. Our results show that metformin prevents or slows down the onset of renal inflammation and fibrosis in mice with UUO, an effect that could be mediated by activation of AMPK.
Collapse
Affiliation(s)
- Rita C Cavaglieri
- Department of Medicine/Nephrology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Robert T Day
- Department of Medicine/Nephrology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Denis Feliers
- Department of Medicine/Nephrology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA.
| | - Hanna E Abboud
- Department of Medicine/Nephrology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
46
|
Prochazkova J, Slavik L, Ulehlova J, Prochazka M. The role of tissue factor in normal pregnancy and in the development of preeclampsia: A review. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2015; 159:192-6. [DOI: 10.5507/bp.2014.061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 11/13/2014] [Indexed: 11/23/2022] Open
|
47
|
Chen X, Walther FJ, Sengers RMA, Laghmani EH, Salam A, Folkerts G, Pera T, Wagenaar GTM. Metformin attenuates hyperoxia-induced lung injury in neonatal rats by reducing the inflammatory response. Am J Physiol Lung Cell Mol Physiol 2015; 309:L262-70. [PMID: 26047641 DOI: 10.1152/ajplung.00389.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 06/02/2015] [Indexed: 01/31/2023] Open
Abstract
Because therapeutic options are lacking for bronchopulmonary dysplasia (BPD), there is an urgent medical need to discover novel targets/drugs to treat this neonatal chronic lung disease. Metformin, a drug commonly used to lower blood glucose in type 2 diabetes patients, may be a novel therapeutic option for BPD by reducing pulmonary inflammation and fibrosis and improving vascularization. We investigated the therapeutic potential of daily treatment with 25 and 100 mg/kg metformin, injected subcutaneously in neonatal Wistar rats with severe experimental BPD, induced by continuous exposure to 100% oxygen for 10 days. Parameters investigated included survival, lung and heart histopathology, pulmonary fibrin and collagen deposition, vascular leakage, right ventricular hypertrophy, and differential mRNA expression in the lungs of key genes involved in BPD pathogenesis, including inflammation, coagulation, and alveolar development. After daily metformin treatment rat pups with experimental BPD had reduced mortality, alveolar septum thickness, lung inflammation, and fibrosis, demonstrated by a reduced influx of macrophages and neutrophils and hyperoxia-induced collagen III and fibrin deposition (25 mg/kg), as well as improved vascularization (100 mg/kg) compared with control treatment. However, metformin did not ameliorate alveolar enlargement, small arteriole wall thickening, vascular alveolar leakage, and right ventricular hypertrophy. In conclusion metformin prolongs survival and attenuates pulmonary injury by reducing pulmonary inflammation, coagulation, and fibrosis but does not affect alveolar development or prevent pulmonary arterial hypertension and right ventricular hypertrophy in neonatal rats with severe hyperoxia-induced experimental BPD.
Collapse
Affiliation(s)
- Xueyu Chen
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frans J Walther
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands; Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-University of California, Los Angeles Medical Center, Torrance, California
| | - Rozemarijn M A Sengers
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - El Houari Laghmani
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Asma Salam
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gert Folkerts
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands; and
| | - Tonio Pera
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerry T M Wagenaar
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands;
| |
Collapse
|
48
|
Hattori Y, Hattori K, Hayashi T. Pleiotropic benefits of metformin: macrophage targeting its anti-inflammatory mechanisms. Diabetes 2015; 64:1907-9. [PMID: 25999535 DOI: 10.2337/db15-0090] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Yuichi Hattori
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Kohshi Hattori
- Anesthesiology and Pain Relief Center, University of Tokyo Hospital, Tokyo, Japan
| | - Toshio Hayashi
- Department of Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
49
|
Casey SC, Vaccari M, Al-Mulla F, Al-Temaimi R, Amedei A, Barcellos-Hoff MH, Brown DG, Chapellier M, Christopher J, Curran CS, Forte S, Hamid RA, Heneberg P, Koch DC, Krishnakumar PK, Laconi E, Maguer-Satta V, Marongiu F, Memeo L, Mondello C, Raju J, Roman J, Roy R, Ryan EP, Ryeom S, Salem HK, Scovassi AI, Singh N, Soucek L, Vermeulen L, Whitfield JR, Woodrick J, Colacci A, Bisson WH, Felsher DW. The effect of environmental chemicals on the tumor microenvironment. Carcinogenesis 2015; 36 Suppl 1:S160-S183. [PMID: 26106136 PMCID: PMC4565612 DOI: 10.1093/carcin/bgv035] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 11/24/2014] [Accepted: 11/28/2014] [Indexed: 12/11/2022] Open
Abstract
Potentially carcinogenic compounds may cause cancer through direct DNA damage or through indirect cellular or physiological effects. To study possible carcinogens, the fields of endocrinology, genetics, epigenetics, medicine, environmental health, toxicology, pharmacology and oncology must be considered. Disruptive chemicals may also contribute to multiple stages of tumor development through effects on the tumor microenvironment. In turn, the tumor microenvironment consists of a complex interaction among blood vessels that feed the tumor, the extracellular matrix that provides structural and biochemical support, signaling molecules that send messages and soluble factors such as cytokines. The tumor microenvironment also consists of many host cellular effectors including multipotent stromal cells/mesenchymal stem cells, fibroblasts, endothelial cell precursors, antigen-presenting cells, lymphocytes and innate immune cells. Carcinogens can influence the tumor microenvironment through effects on epithelial cells, the most common origin of cancer, as well as on stromal cells, extracellular matrix components and immune cells. Here, we review how environmental exposures can perturb the tumor microenvironment. We suggest a role for disrupting chemicals such as nickel chloride, Bisphenol A, butyltins, methylmercury and paraquat as well as more traditional carcinogens, such as radiation, and pharmaceuticals, such as diabetes medications, in the disruption of the tumor microenvironment. Further studies interrogating the role of chemicals and their mixtures in dose-dependent effects on the tumor microenvironment could have important general mechanistic implications for the etiology and prevention of tumorigenesis.
Collapse
Affiliation(s)
- Stephanie C Casey
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA 94305, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy, Department of Pathology, Kuwait University, 13110 Safat, Kuwait, Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy, Department of Radiation Oncology, NYU School of Medicine, New York, NY 10016, USA, Department of Environmental and Radiological Health Sciences, Colorado State University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Centre De Recherche En Cancerologie De Lyon, U1052-UMR5286, Université de Lyon, 69007 Lyon, France, Cancer Research UK, Cambridge Institute, University of Cambridge, Robinson Way, CB2 0RE Cambridge, UK, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia, Charles University in Prague, Third Faculty of Medicine, 100 00 Prague 10, Czech Republic, Center for Environment and Water, Research Institute, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia, Department of Science and Biomedical Technology, University of Cagliari, 09124 Cagliari, Italy, Pathology Unit, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy, Regulatory Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Department of Medicine, University of Louisville, Louisville, KY 40202, USA, Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA, University of Pennsylvania School of Medicine
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, 13110 Safat, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy
| | | | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences, Colorado State University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Marion Chapellier
- Centre De Recherche En Cancerologie De Lyon, U1052-UMR5286, Université de Lyon, 69007 Lyon, France
| | - Joseph Christopher
- Cancer Research UK, Cambridge Institute, University of Cambridge, Robinson Way, CB2 0RE Cambridge, UK
| | - Colleen S Curran
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Roslida A Hamid
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia
| | - Petr Heneberg
- Charles University in Prague, Third Faculty of Medicine, 100 00 Prague 10, Czech Republic
| | - Daniel C Koch
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA 94305, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy, Department of Pathology, Kuwait University, 13110 Safat, Kuwait, Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy, Department of Radiation Oncology, NYU School of Medicine, New York, NY 10016, USA, Department of Environmental and Radiological Health Sciences, Colorado State University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Centre De Recherche En Cancerologie De Lyon, U1052-UMR5286, Université de Lyon, 69007 Lyon, France, Cancer Research UK, Cambridge Institute, University of Cambridge, Robinson Way, CB2 0RE Cambridge, UK, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia, Charles University in Prague, Third Faculty of Medicine, 100 00 Prague 10, Czech Republic, Center for Environment and Water, Research Institute, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia, Department of Science and Biomedical Technology, University of Cagliari, 09124 Cagliari, Italy, Pathology Unit, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy, Regulatory Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Department of Medicine, University of Louisville, Louisville, KY 40202, USA, Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA, University of Pennsylvania School of Medicine
| | - P K Krishnakumar
- Center for Environment and Water, Research Institute, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| | - Ezio Laconi
- Department of Science and Biomedical Technology, University of Cagliari, 09124 Cagliari, Italy
| | - Veronique Maguer-Satta
- Centre De Recherche En Cancerologie De Lyon, U1052-UMR5286, Université de Lyon, 69007 Lyon, France
| | - Fabio Marongiu
- Department of Science and Biomedical Technology, University of Cagliari, 09124 Cagliari, Italy
| | - Lorenzo Memeo
- Pathology Unit, Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Jayadev Raju
- Regulatory Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Jesse Roman
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Sandra Ryeom
- University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Hosni K Salem
- Urology Department, Kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 11562, Egypt
| | - A Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Neetu Singh
- Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India
| | - Laura Soucek
- Vall d'Hebron Institute of Oncology (VHIO) and Institució Catalana de Recerca i Estudis Avançats (ICREA), 08035 Barcelona, Spain
| | - Louis Vermeulen
- Center for Experimental Molecular Medicine (CEMM), Academic Medical Center (AMC), Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jonathan R Whitfield
- Vall d'Hebron Institute of Oncology (VHIO) and Institució Catalana de Recerca i Estudis Avançats (ICREA), 08035 Barcelona, Spain
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - William H Bisson
- Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA, and
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
50
|
Abstract
Improved treatments are needed for nearly all forms of Mycobacterium tuberculosis infection. Adjunctive host-directed therapies have the potential to shorten tuberculosis treatment duration, prevent resistance and reduce lung injury by promoting autophagy, antimicrobial peptide production and other macrophage effector mechanisms, as well as by modifying specific mechanisms that cause lung inflammation and matrix destruction. The range of candidates is broad, including several agents approved for other clinical indications that are ready for evaluation in Phase II clinical trials. The promise of new and existing host-directed therapies that could accelerate response and improve tuberculosis treatment outcomes is discussed in this Opinion article.
Collapse
|