1
|
Barkas GI, Karakousis ND, Gourgoulianis KI, Daniil Z, Papanas N, Kotsiou OS. Pioglitazone and asthma: a review of current evidence. J Asthma 2025; 62:365-375. [PMID: 39373513 DOI: 10.1080/02770903.2024.2414342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/19/2024] [Accepted: 10/05/2024] [Indexed: 10/08/2024]
Abstract
OBJECTIVE This review aims to present existing evidence on the impact of pioglitazone, a thiazolidinedione class anti-diabetic drug, on asthma control and lung function, providing a comprehensive understanding of its potential as a treatment for asthma. DATA SOURCES The review draws upon data from preclinical animal studies and clinical trials investigating the effects of pioglitazone on asthma, focusing on its role in reducing airway inflammation, hyperreactivity, and remodeling, and its impact on pulmonary function. STUDY SELECTIONS Relevant studies were selected based on their examination of pioglitazone's therapeutic effects in asthma, including both animal models and clinical trials involving human asthma patients. RESULTS Animal studies have suggested that pioglitazone could alleviate inflammation, airway hyperreactivity, and airway remodeling, thereby improving pulmonary function in asthma. However, clinical trials have not demonstrated significant therapeutic benefits, with minimal improvements observed in asthma control and lung function, and the presence of notable side effects. CONCLUSION Despite promising preclinical data, the efficacy of pioglitazone in treating human asthma remains unproven, with safety concerns and limited clinical benefits observed in trials. Further research is needed to assess the safety and effectiveness of pioglitazone in asthma treatment and to explore its impact on other inflammatory mechanisms.
Collapse
Affiliation(s)
- Georgios I Barkas
- Human Pathophysiology Department, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Nikolaos D Karakousis
- Department of Respiratory Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
- Department of Nursing, School of Health Sciences, University of Thessaly, Larissa, Greece
| | | | - Zoe Daniil
- Department of Respiratory Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Nikolaos Papanas
- Second Department of Internal Medicine, Diabetes Center-Diabetic Foot Clinic, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ourania S Kotsiou
- Human Pathophysiology Department, School of Health Sciences, University of Thessaly, Larissa, Greece
- Department of Respiratory Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
2
|
Beigoli S, Kiani S, Asgharzadeh F, Memarzia A, Boskabady MH. Promising role of peroxisome proliferator-activated receptors in respiratory disorders, a review. Drug Metab Rev 2025; 57:26-50. [PMID: 39726246 DOI: 10.1080/03602532.2024.2442012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
Several studies indicate various pharmacological and therapeutic effects of peroxisome proliferator-activated receptors (PPARs) in different disorders. The current review describes the influences of PPARs on respiratory, allergic, and immunologic diseases. Various databases, including PubMed, Science Direct, and Scopus, were searched regarding the effect of PPARs on respiratory and allergic disorders from 1990 to 2024. The effects of PPARs stimulation on experimental animal models of respiratory diseases such as asthma, chronic obstructive pulmonary diseases (COPD), pulmonary fibrosis (PF), and lung infections were shown. Therapeutic potential mediated through PPARs has also been demonstrated in lung cancer, lung infections, and allergic and immunologic disorders. However, few clinical studies showed PPARs mediated therapeutic effects on asthma and COPD. The PPARs-mediated effects on various respiratory disorders were shown through antioxidant, immunomodulatory, anti-inflammatory, and other mechanisms. Therefore, this review indicated possible remedy effects mediated by these receptors in treating respiratory, allergic, and immunologic diseases. Moreover, this mechanistic review paves the way for researchers to consider further experimental and clinical studies.
Collapse
Affiliation(s)
- Sima Beigoli
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Kiani
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, ROYAN Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Brain and Cognitive Sciences, Cell Science Research Center, ROYAN Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fereshteh Asgharzadeh
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arghavan Memarzia
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Hossein Boskabady
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Noori M, Azimirad M, Ghorbaninejad M, Meyfour A, Zali MR, Yadegar A. PPAR-γ agonist mitigates intestinal barrier dysfunction and inflammation induced by Clostridioides difficile SlpA in vitro. Sci Rep 2024; 14:32087. [PMID: 39738433 PMCID: PMC11686163 DOI: 10.1038/s41598-024-83815-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
Clostridioides difficile is the leading cause of healthcare- and antibiotic-associated diarrhea. Surface layer protein A (SlpA), an essential component of the bacterium's outermost layer, contributes to colonization and inflammation. The peroxisome proliferator-activated receptor gamma (PPAR-γ) has been demonstrated to improve intestinal integrity and prevent inflammation in host cells. Here, we investigated the role of PPAR-γ in SlpA-mediated inflammation in Caco-2 cells and THP-1 derived macrophages. The extraction of SlpA was carried out for three toxigenic C. difficile clinical strains (RT126, RT001, RT084) and a non-toxigenic strain (ATCC 700057). The gene expression of tight junction (TJ) proteins and inflammatory markers was determined using RT-qPCR. The production of proinflammatory cytokines and nitric oxide was measured by ELISA and Griss reaction, respectively. Western blotting was performed to detect PPAR-γ level before and after adding its agonist, pioglitazone. SlpA of C. difficile strains enhanced the expression of TLR-4, NF-κB, MyD88, IL-17, MCP-1, IL-8, IL-6, TNF-α, IL-1β, whilst the gene expression level of JAM-A, claudin-1, occludin, PPAR-γ and its receptor (CD36) was decreased in both Caco-2 cells and THP-1 derived macrophages. Moreover, pioglitazone caused a notable elevation in the expression level of PPAR-γ, only following treatment with RT126 SlpA. Besides, pioglitazone pretreatment improved TJ impairment in Caco-2 cells and attenuated proinflammatory cytokine expression in both SlpA-treated cell lines. SlpA can attenuate PPAR-γ expression, trigger TJ disruption, and stimulate inflammatory response in host cells. Notably, these events could be reversed by pretreatment of cells with PPAR-γ agonist. Further experiments are required to corroborate the present findings.
Collapse
Affiliation(s)
- Maryam Noori
- Foodborne and Waterborne Diseases Research Center , Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Azimirad
- Foodborne and Waterborne Diseases Research Center , Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Ghorbaninejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anna Meyfour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center , Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Kathirasan DRAL, Normizan SN'IB, Salleh NABM, Poh-Yen K. Recent progress on small molecule TLR4 antagonist against triple-negative breast cancer progression and complications. Bioorg Med Chem 2024; 116:118000. [PMID: 39561584 DOI: 10.1016/j.bmc.2024.118000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/02/2024] [Accepted: 11/08/2024] [Indexed: 11/21/2024]
Abstract
Toll-like receptor 4 (TLR4) plays a vital role in the innate immune response, but its overactivation has been associated with several diseases, such as aggressive progression of triple-negative breast cancer (TNBC). As a result, inhibiting TLR4 has emerged as a potential therapeutic strategy for this challenging breast cancer subtype. This review summarizes recent advancements in the development of small-molecule TLR4 antagonists to suppress TNBC growth, metastasis, and chemotherapy resistance. We also examine their potential in managing cancer-related complications and propose future directions for their application in TNBC therapy.
Collapse
Affiliation(s)
- Darsshen Ramana A L Kathirasan
- Faculty Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, 30450 Ipoh, Perak, Malaysia; Pharmacy Department, Hospital Raja Permaisuri Bainun, Jalan Raja Ashman Shah, 30450 Ipoh, Perak, Malaysia
| | - Siti Nor 'Izzah Binti Normizan
- Faculty Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, 30450 Ipoh, Perak, Malaysia
| | | | - Khor Poh-Yen
- Faculty Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, 30450 Ipoh, Perak, Malaysia.
| |
Collapse
|
5
|
Pérez-Villalobos MC, Barba-González A, García-Carrillo N, Muñoz-Ortega MH, Sánchez-Alemán E, Ávila-Blanco ME, Morones-Gamboa JC, Ventura-Juárez J, Martínez-Hernández SL. Nephroprotective effect of pioglitazone in a Wistar rat model of adenine‑induced chronic kidney disease. Exp Ther Med 2024; 28:392. [PMID: 39161617 PMCID: PMC11332140 DOI: 10.3892/etm.2024.12681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/27/2024] [Indexed: 08/21/2024] Open
Abstract
Chronic kidney disease (CKD) is a progressive disease with a high mortality rate and a worldwide prevalence of 13.4%, triggered by various diseases with high incidence. The aim of the present study was to investigate the anti-inflammatory and antifibrotic effect of pioglitazone on kidney in an adenine-induced Wistar rats and the mechanisms possibly involved. CKD was induced in 40 rats. Rats were divided into two groups, which were split into the following sub-groups: i) Therapeutic (pioglitazone administered after renal damage) divided into intact (healthy), adenine (CKD) and adenine/pioglitazone (treatment) and ii) prophylactic (adenine and pioglitazone administered at the same time) split into intact (healthy), adenine (CKD), endogenous reversion (recovery without treatment), adenine/pioglitazone (treatment) and pioglitazone sub-groups. Reverse transcription-quantitative PCR (collagen I, α-SMA and TGF-β), and hematoxylin-eosin, Masson's trichrome and Sirius red staining were performed to measure histological markers of kidney damage, also the serum markers (urea, creatinine and uric acid) were performed, for analyze the effects of pioglitazone. In the adenine/pioglitazone rats of the therapeutic group, renal function parameters such as eGFR increased and serum creatinine decreased from those of untreated rats (CKD), however the renal index, serum urea, abnormalities in renal morphology, inflammatory cells and relative gene expression of collagen I, α-SMA and TGF-β did not change relative to the CKD rats. In adenine/pioglitazone rats, extracellular matrix collagen accumulation was significantly lower than the CKD rats. On the other hand, in adenine/pioglitazone rats of the prophylactic group, the renal index, creatinine, urea, uric acid serum and relative gene expression of collagen I, α-SMA, and TGF-β were significantly lower, as well as the presence of 2,8-dihydroxyadenine crystals, and extracellular matrix collagen compared with CKD rats. In addition, the eGFR in the treatment group was similar to healthy rats, renal morphology was restored, and inflammatory cells were significantly lower. In conclusion, pioglitazone has a nephroprotective effect when administered in the early stages of kidney damage, reducing inflammatory and fibrotic processes and improving glomerular filtration rate. Furthermore, in the late phase of treatment, a tendency to decrease creatinine and increase eGFR was observed.
Collapse
Affiliation(s)
| | - Andrea Barba-González
- Department of Morphology, Center of Basic Sciences, Autonomous University of Aguascalientes, 20100 Aguascalientes, Mexico
| | - Nicté García-Carrillo
- Department of Morphology, Center of Basic Sciences, Autonomous University of Aguascalientes, 20100 Aguascalientes, Mexico
| | - Martín Humberto Muñoz-Ortega
- Department of Chemistry, Center of Basic Sciences, Autonomous University of Aguascalientes, 20100 Aguascalientes, Mexico
| | - Esperanza Sánchez-Alemán
- Department of Morphology, Center of Basic Sciences, Autonomous University of Aguascalientes, 20100 Aguascalientes, Mexico
- Family Medicine Unit 8, Mexican Social Security Institute, 20180 Aguascalientes, Mexico
| | - Manuel Enrique Ávila-Blanco
- Department of Morphology, Center of Basic Sciences, Autonomous University of Aguascalientes, 20100 Aguascalientes, Mexico
| | | | - Javier Ventura-Juárez
- Department of Morphology, Center of Basic Sciences, Autonomous University of Aguascalientes, 20100 Aguascalientes, Mexico
| | - Sandra Luz Martínez-Hernández
- Department of Microbiology, Center of Basic Sciences, Autonomous University of Aguascalientes, 20100 Aguascalientes, Mexico
| |
Collapse
|
6
|
Baagar K, Alessa T, Abu-Farha M, Abubaker J, Alhumaidi H, Franco Ceruto JA, Hamad MK, Omrani A, Abdelrahman S, Zaka-Ul Haq M, Safi AW, Alhariri B, Barman M, Abdelmajid A, Cancio HVD, Elmekaty E, Al-Khairi I, Cherian P, Jayyousi L, Ahmed M, Qaddoumi M, Hajji S, Esmaeel A, Al-Andaleeb A, Channanath A, Devarajan S, Ali H, Thanaraj TA, Al-Sabah S, Al-Mulla F, Abdul-Ghani M, Jayyousi A. Effect of pioglitazone on inflammatory response and clinical outcome in T2DM patients with COVID-19: a randomized multicenter double-blind clinical trial. Front Immunol 2024; 15:1369918. [PMID: 39308871 PMCID: PMC11412854 DOI: 10.3389/fimmu.2024.1369918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) caused by the coronavirus SARS-CoV-2, has emerged as a rapidly spreading contagious disease across the globe. Recent studies showed that people with diabetes mellitus, severe obesity, and cardiovascular disease are at higher risk of mortality from COVID-19. It has been suggested that the increased risk is due to the chronic inflammatory state associated with type 2 diabetes. This study aimed to evaluate the efficacy of pioglitazone, a strong insulin sensitizer with anti-inflammatory properties, in improving the clinical outcomes of patients with type 2 diabetes admitted with moderate-severe COVID-19. Method We enrolled 350 patients with type 2 diabetes who were admitted to hospitals in Qatar and Kuwait with COVID-19. Patients were randomized to receive, in a double-blind fashion, pioglitazone (n = 189) or a matching placebo (n = 161) for 28 days. The study had two primary outcomes: (1) the incidence of a composite outcome composed of (a) the requirement for mechanical ventilation, (b) death, and (c) myocardial damage; and (2) an increase in C-reactive protein (CRP) levels. Results The first primary outcome occurred in 28 participants (8%), and the secondary outcome occurred in 17. Treatment with pioglitazone showed a significant reduction in interleukin (IL)-3 levels compared with placebo treatment (mean (SD) 2.73 (± 2.14) [95% CI: 0.02, 1.1], p = 0.043 vs. 2.28 (± 1.67) [95% CI: - 0.23, 0.86], p = 0.3, respectively), with no effect seen in the levels of other inflammatory markers. Even though not significant, a few of the patients on pioglitazone exhibited serum troponin levels > 3 times higher than the normal range seen in patients on placebo. On the other hand, more patients on pioglitazone were admitted to the ICU than those with placebo, and no significant difference in the CRP reduction was observed between the two groups. Conclusion The results of the present study demonstrate that pioglitazone treatment did not independently provide any additional clinical benefit to patients with type 2 diabetes admitted with a COVID-19 infection. Clinical trial registration https://clinicaltrials.gov, identifier NCT04604223.
Collapse
Affiliation(s)
- Khaled Baagar
- Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | - Thamer Alessa
- Jaber AlAhmed Hospital, Ministry of Health, Kuwait City, Kuwait
| | - Mohamed Abu-Farha
- Department of Translational Research, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Jehad Abubaker
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Heba Alhumaidi
- Jaber AlAhmed Hospital, Ministry of Health, Kuwait City, Kuwait
| | | | | | - Ali Omrani
- Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | | | | | | | | | - Manish Barman
- Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | | | | | - Eman Elmekaty
- Pharmacy Department, Hamad Medical Corporation, Doha, Qatar
| | - Irina Al-Khairi
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Preethi Cherian
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Lina Jayyousi
- Royal College of Surgeons in Ireland, Medical University of Bahrain, Manama, Bahrain
| | - Mohammed Ahmed
- Department of Translational Research, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Mohammed Qaddoumi
- Pharmacology and Therapeutics Department, Faculty of Pharmacy, Kuwait University, Kuwait City, Kuwait
| | - Sulaiman Hajji
- Jaber AlAhmed Hospital, Ministry of Health, Kuwait City, Kuwait
| | - Ahmad Esmaeel
- Jaber AlAhmed Hospital, Ministry of Health, Kuwait City, Kuwait
| | - Ali Al-Andaleeb
- Jaber AlAhmed Hospital, Ministry of Health, Kuwait City, Kuwait
| | | | | | - Hamad Ali
- Dasman Diabetes Institute, Kuwait City, Kuwait
| | | | - Salman Al-Sabah
- Jaber AlAhmed Hospital, Ministry of Health, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Department of Translational Research, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Muhammad Abdul-Ghani
- Department of Translational Research, Dasman Diabetes Institute, Kuwait City, Kuwait
- Division of Diabetes, University of Texas Health Science Center, San Antonio, TX, United States
| | - Amin Jayyousi
- Medicine Department, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
7
|
Tang J, Dong W, Wang D, Deng Q, Guo H, Xiao G. Upregulation of PGC-1α expression by pioglitazone mediates prevention of sepsis-induced acute lung injury. Braz J Med Biol Res 2024; 57:e13235. [PMID: 38511769 PMCID: PMC10946242 DOI: 10.1590/1414-431x2024e13235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/25/2024] [Indexed: 03/22/2024] Open
Abstract
The imbalance between pro-inflammatory M1 and anti-inflammatory M2 macrophages plays a critical role in the pathogenesis of sepsis-induced acute lung injury (ALI). Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) may modulate macrophage polarization toward the M2 phenotype by altering mitochondrial activity. This study aimed to investigate the role of the PGC-1α agonist pioglitazone (PGZ) in modulating sepsis-induced ALI. A mouse model of sepsis-induced ALI was established using cecal ligation and puncture (CLP). An in vitro model was created by stimulating MH-S cells with lipopolysaccharide (LPS). qRT-PCR was used to measure mRNA levels of M1 markers iNOS and MHC-II and M2 markers Arg1 and CD206 to evaluate macrophage polarization. Western blotting detected expression of peroxisome proliferator-activated receptor gamma (PPARγ) PGC-1α, and mitochondrial biogenesis proteins NRF1, NRF2, and mtTFA. To assess mitochondrial content and function, reactive oxygen species levels were detected by dihydroethidium staining, and mitochondrial DNA copy number was measured by qRT-PCR. In the CLP-induced ALI mouse model, lung tissues exhibited reduced PGC-1α expression. PGZ treatment rescued PGC-1α expression and alleviated lung injury, as evidenced by decreased lung wet-to-dry weight ratio, pro-inflammatory cytokine secretion (tumor necrosis factor-α, interleukin-1β, interleukin-6), and enhanced M2 macrophage polarization. Mechanistic investigations revealed that PGZ activated the PPARγ/PGC-1α/mitochondrial protection pathway to prevent sepsis-induced ALI by inhibiting M1 macrophage polarization. These results may provide new insights and evidence for developing PGZ as a potential ALI therapy.
Collapse
Affiliation(s)
- Jing Tang
- Department of Infectious Diseases, Ziyang First People's Hospital, Ziyang, China
| | - Wenzhu Dong
- Department of Infectious Diseases, Ziyang First People's Hospital, Ziyang, China
| | - Dan Wang
- Department of Infectious Diseases, Ziyang First People's Hospital, Ziyang, China
| | - Qin Deng
- Department of Infectious Diseases, Ziyang First People's Hospital, Ziyang, China
| | - Honggang Guo
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou, China
| | - Guibao Xiao
- Department of Infectious Diseases, Ziyang First People's Hospital, Ziyang, China
| |
Collapse
|
8
|
Ertuglu LA, Deger SM, Alsouqi A, Hung A, Gamboa J, Mambungu C, Sha F, Siew E, Abumrad NN, Ikizler TA. A randomized controlled pilot trial of anakinra and pioglitazone for protein metabolism in patients on maintenance haemodialysis. J Cachexia Sarcopenia Muscle 2024; 15:401-411. [PMID: 38178557 PMCID: PMC10834322 DOI: 10.1002/jcsm.13395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/17/2023] [Accepted: 11/02/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Chronic inflammation and insulin resistance are highly prevalent in patients on maintenance haemodialysis (MHD) and are strongly associated with protein energy wasting. We conducted a pilot, randomized, placebo-controlled trial of recombinant human interleukin-1 receptor antagonist (IL-1ra) and pioglitazone to explore the safety, feasibility and efficacy for insulin-mediated protein metabolism in patients undergoing MHD. METHODS Twenty-four patients were randomized to receive IL-1ra, pioglitazone or placebo for 12 weeks. Changes in serum inflammatory markers and insulin-mediated protein synthesis, breakdown and net balance in the whole-body and skeletal muscle compartments were assessed using hyperinsulinaemic-hyperaminoacidemic clamp technique at baseline and Week 12. RESULTS Among 24 patients, median (interquartile range) age was 51 (40, 61), 79% were African American and 21% had diabetes mellitus. All patients initiated on intervention completed the study, and no serious adverse events were observed. There was a statistically significant decrease in serum high-sensitivity C-reactive protein in the pioglitazone group compared with placebo, but not in the IL-1ra group. No significant differences in the changes of whole-body or skeletal muscle protein synthesis, breakdown and net balance were found between the groups. CONCLUSIONS In this pilot study, there were no statistically significant effects of 12 weeks of IL-1ra or pioglitazone on protein metabolism in patients on MHD. CLINICALTRIALS gov registration: NCT02278562.
Collapse
Affiliation(s)
- Lale A Ertuglu
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Serpil Muge Deger
- Department of Nephrology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Aseel Alsouqi
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
- Now with Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Adriana Hung
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Administration Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Jorge Gamboa
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cindy Mambungu
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Feng Sha
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Edward Siew
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Administration Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Naji N Abumrad
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - T Alp Ikizler
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Administration Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
9
|
Othman AM, Ashour Ibrahim I, Saleh SM, Abo-Elmatty DM, Mesbah NM, Abdel-Hamed AR. The Safety and Efficacy of Combining Saxagliptin and Pioglitazone Therapy in Streptozocin-Induced Diabetic Rats. Biomedicines 2023; 11:3300. [PMID: 38137521 PMCID: PMC10741989 DOI: 10.3390/biomedicines11123300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a chronic progressive disease due to insulin resistance. Oxidative stress complicates the etiology of T2DM. Saxagliptin is a selective dipeptidyl peptidase-4 (DPP-4) inhibitor, while Pioglitazone is a thiazolidinedione insulin sensitizer. This study aimed to assess the effect of Saxagliptin and Pioglitazone monotherapy and combination therapy on the biochemical and biological parameters in streptozotocin (STZ)-induced diabetic rats. METHODS The study included thirty-five male albino rats. Diabetes mellitus was induced by intraperitoneal STZ injection (35 mg/kg). For a 1-month duration, rats were divided into five groups. Glucose homeostasis traits, lipid profiles, kidney functions, liver enzymes, and oxidative stress markers were measured. Gene expression of miRNA-29a, phosphoenolpyruvate carboxykinase (PEPCK), phosphoinositide-3-kinase (PI3K), and interleukin 1 beta (IL-1β) was assessed using qRT-PCR. RESULTS At a 1-month treatment duration, combination therapy improves oxidative stress markers more than either drug alone. The combination therapy had significantly higher levels of SOD, catalase, and GSH and lower levels of MDA compared to the monotherapy. Additionally, the diabetic group showed a significant increase in the expression levels of miRNA-29a, PEPCK, and IL-1β and a significant decrease in PI3K compared to the normal control group. However, combination therapy of Saxagliptin and Pioglitazone was more effective than either Saxagliptin or Pioglitazone alone in reversing these results, especially for PEPCK and IL-1β. CONCLUSIONS Our findings revealed that combining Saxagliptin and Pioglitazone improves glycemic control and genetic and epigenetic expression profiles, which play an essential regulatory role in normal metabolism.
Collapse
Affiliation(s)
- Ahmed Mohamed Othman
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt (N.M.M.)
| | - Ibrahim Ashour Ibrahim
- Department of Biochemistry, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Samy M. Saleh
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt (N.M.M.)
| | - Dina M. Abo-Elmatty
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt (N.M.M.)
| | - Noha M. Mesbah
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt (N.M.M.)
| | - Asmaa R. Abdel-Hamed
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt (N.M.M.)
| |
Collapse
|
10
|
Lira Chavez FM, Gartzke LP, van Beuningen FE, Wink SE, Henning RH, Krenning G, Bouma HR. Restoring the infected powerhouse: Mitochondrial quality control in sepsis. Redox Biol 2023; 68:102968. [PMID: 38039825 PMCID: PMC10711241 DOI: 10.1016/j.redox.2023.102968] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023] Open
Abstract
Sepsis is a dysregulated host response to an infection, characterized by organ failure. The pathophysiology is complex and incompletely understood, but mitochondria appear to play a key role in the cascade of events that culminate in multiple organ failure and potentially death. In shaping immune responses, mitochondria fulfil dual roles: they not only supply energy and metabolic intermediates crucial for immune cell activation and function but also influence inflammatory and cell death pathways. Importantly, mitochondrial dysfunction has a dual impact, compromising both immune system efficiency and the metabolic stability of end organs. Dysfunctional mitochondria contribute to the development of a hyperinflammatory state and loss of cellular homeostasis, resulting in poor clinical outcomes. Already in early sepsis, signs of mitochondrial dysfunction are apparent and consequently, strategies to optimize mitochondrial function in sepsis should not only prevent the occurrence of mitochondrial dysfunction, but also cover the repair of the sustained mitochondrial damage. Here, we discuss mitochondrial quality control (mtQC) in the pathogenesis of sepsis and exemplify how mtQC could serve as therapeutic target to overcome mitochondrial dysfunction. Hence, replacing or repairing dysfunctional mitochondria may contribute to the recovery of organ function in sepsis. Mitochondrial biogenesis is a process that results in the formation of new mitochondria and is critical for maintaining a pool of healthy mitochondria. However, exacerbated biogenesis during early sepsis can result in accumulation of structurally aberrant mitochondria that fail to restore bioenergetics, produce excess reactive oxygen species (ROS) and exacerbate the disease course. Conversely, enhancing mitophagy can protect against organ damage by limiting the release of mitochondrial-derived damage-associated molecules (DAMPs). Furthermore, promoting mitophagy may facilitate the growth of healthy mitochondria by blocking the replication of damaged mitochondria and allow for post sepsis organ recovery through enabling mitophagy-coupled biogenesis. The remaining healthy mitochondria may provide an undamaged scaffold to reproduce functional mitochondria. However, the kinetics of mtQC in sepsis, specifically mitophagy, and the optimal timing for intervention remain poorly understood. This review emphasizes the importance of integrating mitophagy induction with mtQC mechanisms to prevent undesired effects associated with solely the induction of mitochondrial biogenesis.
Collapse
Affiliation(s)
- F M Lira Chavez
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands.
| | - L P Gartzke
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands
| | - F E van Beuningen
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands
| | - S E Wink
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands
| | - R H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands
| | - G Krenning
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands; Sulfateq B.V, Admiraal de Ruyterlaan 5, 9726, GN Groningen, the Netherlands
| | - H R Bouma
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands; Department of Internal Medicine, University Medical Centre Groningen, University of Groningen, 9713, GZ Groningen, the Netherlands
| |
Collapse
|
11
|
Victor S, Forbes B, Greenough A, Edwards AD. PPAR Gamma Receptor: A Novel Target to Improve Morbidity in Preterm Babies. Pharmaceuticals (Basel) 2023; 16:1530. [PMID: 38004396 PMCID: PMC10675178 DOI: 10.3390/ph16111530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/19/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Worldwide, three-quarters of a million babies are born extremely preterm (<28 weeks gestation) with devastating outcomes: 20% die in the newborn period, a further 35% develop bronchopulmonary dysplasia (BPD), and 10% suffer from cerebral palsy. Pioglitazone, a Peroxisome Proliferator Activated Receptor Gamma (PPARγ) agonist, may reduce the incidence of BPD and improve neurodevelopment in extreme preterm babies. Pioglitazone exerts an anti-inflammatory action mediated through Nuclear Factor-kappa B repression. PPARγ signalling is underactive in preterm babies as adiponectin remains low during the neonatal period. In newborn animal models, pioglitazone has been shown to be protective against BPD, necrotising enterocolitis, and lipopolysaccharide-induced brain injury. Single Nucleotide Polymorphisms of PPARγ are associated with inhibited preterm brain development and impaired neurodevelopment. Pioglitazone was well tolerated by the foetus in reproductive toxicology experiments. Bladder cancer, bone fractures, and macular oedema, seen rarely in adults, may be avoided with a short treatment course. The other effects of pioglitazone, including improved glycaemic control and lipid metabolism, may provide added benefit in the context of prematurity. Currently, there is no formulation of pioglitazone suitable for administration to preterm babies. A liquid formulation of pioglitazone needs to be developed before clinical trials. The potential benefits are likely to outweigh any anticipated safety concerns.
Collapse
Affiliation(s)
- Suresh Victor
- Centre for the Developing Brain, Department of Perinatal Imaging and Health, School of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, UK;
| | - Ben Forbes
- Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, Stamford Street, London SE1 9NH, UK;
| | - Anne Greenough
- Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London, Neonatal Intensive Care Centre, Floor 4, Golden Jubilee Wing, King’s College Hospital, Denmark Hill, Brixton, London SE5 9RS, UK;
| | - A. David Edwards
- Centre for the Developing Brain, Department of Perinatal Imaging and Health, School of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, UK;
| |
Collapse
|
12
|
Mukherjee AG, Renu K, Gopalakrishnan AV, Jayaraj R, Dey A, Vellingiri B, Ganesan R. Epicardial adipose tissue and cardiac lipotoxicity: A review. Life Sci 2023; 328:121913. [PMID: 37414140 DOI: 10.1016/j.lfs.2023.121913] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/21/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Epicardial adipose tissue (EAT) has morphological and physiological contiguity with the myocardium and coronary arteries, making it a visceral fat deposit with some unique properties. Under normal circumstances, EAT exhibits biochemical, mechanical, and thermogenic cardioprotective characteristics. Under clinical processes, epicardial fat can directly impact the heart and coronary arteries by secreting proinflammatory cytokines via vasocrine or paracrine mechanisms. It is still not apparent what factors affect this equilibrium. Returning epicardial fat to its physiological purpose may be possible by enhanced local vascularization, weight loss, and focused pharmacological therapies. This review centers on EAT's developing physiological and pathophysiological dimensions and its various and pioneering clinical utilities.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India.
| | - Rama Jayaraj
- Jindal Institute of Behavioral Sciences (JIBS), Jindal Global Institution of Eminence Deemed to Be University, 28, Sonipat 131001, India; Director of Clinical Sciences, Northern Territory Institute of Research and Training, Darwin, NT 0909, Australia
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal 700073, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
13
|
Balkrishna A, Sinha S, Kumar A, Arya V, Gautam AK, Valis M, Kuca K, Kumar D, Amarowicz R. Sepsis-mediated renal dysfunction: Pathophysiology, biomarkers and role of phytoconstituents in its management. Biomed Pharmacother 2023; 165:115183. [PMID: 37487442 DOI: 10.1016/j.biopha.2023.115183] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/08/2023] [Accepted: 07/18/2023] [Indexed: 07/26/2023] Open
Abstract
Sepsis has evolved as an enormous health issue amongst critically ill patients. It is a major risk factor that results in multiple organ failure and shock. Acute kidney injury (AKI) is one of the most frequent complications underlying sepsis, which portends a heavy burden of mortality and morbidity. Thus, the present review is aimed to provide an insight into the recent progression in the molecular mechanisms targeting dysregulated immune response and cellular dysfunction involved in the development of sepsis-associated AKI, accentuating the phytoconstituents as eligible candidates for attenuating the onset and progression of sepsis-associated AKI. The pathogenesis of sepsis-mediated AKI entails a complicated mechanism and is likely to involve a distinct constellation of hemodynamic, inflammatory, and immune mechanisms. Novel biomarkers like neutrophil gelatinase-associated lipocalin, soluble triggering receptor expressed on myeloid cells 1, procalcitonin, alpha-1-microglobulin, and presepsin can help in a more sensitive diagnosis of sepsis-associated AKI. Many bioactive compounds like curcumin, resveratrol, baicalin, quercetin, and polydatin are reported to play an important role in the prevention and management of sepsis-associated AKI by decreasing serum creatinine, blood urea nitrogen, cystatin C, lipid peroxidation, oxidative stress, IL-1β, TNF-α, NF-κB, and increasing the activity of antioxidant enzymes and level of PPARγ. The plant bioactive compounds could be developed into a drug-developing candidate in managing sepsis-mediated acute kidney injury after detailed follow-up studies. Lastly, the gut-kidney axis may be a more promising therapeutic target against the onset of septic AKI, but a deeper understanding of the molecular pathways is still required.
Collapse
Affiliation(s)
- Acharya Balkrishna
- Patanjali Herbal Research Department, Patanjali Research Institute, Haridwar, India
| | - Sugandh Sinha
- Patanjali Herbal Research Department, Patanjali Research Institute, Haridwar, India
| | - Ashwani Kumar
- Patanjali Herbal Research Department, Patanjali Research Institute, Haridwar, India.
| | - Vedpriya Arya
- Patanjali Herbal Research Department, Patanjali Research Institute, Haridwar, India
| | - Ajay Kumar Gautam
- Patanjali Herbal Research Department, Patanjali Research Institute, Haridwar, India
| | - Martin Valis
- Department of Neurology, Charles University in Prague, Faculty of Medicine in Hradec Králové and University Hospital, Hradec Králové, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic; Biomedical Research Center, University Hospital in Hradec Kralove, Sokolska 581, Hradec Kralove, Czech Republic.
| | - Dinesh Kumar
- School of Bioengineering and Food Technology, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Ryszard Amarowicz
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
14
|
Muniz-Santos R, Lucieri-Costa G, de Almeida MAP, Moraes-de-Souza I, Brito MADSM, Silva AR, Gonçalves-de-Albuquerque CF. Lipid oxidation dysregulation: an emerging player in the pathophysiology of sepsis. Front Immunol 2023; 14:1224335. [PMID: 37600769 PMCID: PMC10435884 DOI: 10.3389/fimmu.2023.1224335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/30/2023] [Indexed: 08/22/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by abnormal host response to infection. Millions of people are affected annually worldwide. Derangement of the inflammatory response is crucial in sepsis pathogenesis. However, metabolic, coagulation, and thermoregulatory alterations also occur in patients with sepsis. Fatty acid mobilization and oxidation changes may assume the role of a protagonist in sepsis pathogenesis. Lipid oxidation and free fatty acids (FFAs) are potentially valuable markers for sepsis diagnosis and prognosis. Herein, we discuss inflammatory and metabolic dysfunction during sepsis, focusing on fatty acid oxidation (FAO) alterations in the liver and muscle (skeletal and cardiac) and their implications in sepsis development.
Collapse
Affiliation(s)
- Renan Muniz-Santos
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giovanna Lucieri-Costa
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Matheus Augusto P. de Almeida
- Neuroscience Graduate Program, Federal Fluminense University, Niteroi, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Isabelle Moraes-de-Souza
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Adriana Ribeiro Silva
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Neuroscience Graduate Program, Federal Fluminense University, Niteroi, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
15
|
Jafari-Khataylou Y, Kazemi-Darabadi S, Ahmadi Afshar S. Effects of adenosine N1-Oxide and pioglitazone on inflammatory and antioxidant state in sepsis caused by experimental cecal puncture in rat. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2023; 14:381-387. [PMID: 37564357 PMCID: PMC10410109 DOI: 10.30466/vrf.2022.562229.3625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/12/2022] [Indexed: 08/12/2023]
Abstract
Sepsis is an acute condition caused by the systemic inflammatory response syndrome to an infection. There are very few drugs that could improve the severe conditions in patients with sepsis. Hence, it is important to consider different treatment options. In this survey, we studied the effect of adenosine N1-oxide (ANO) and pioglitazone on rats with cecal ligation and perforation (CLP). They were randomly divided to four groups (n = 10) including Group A: as control group receiving normal saline, Group B: the rats with CLP as surgical control group, Group C: the rats receiving ANO, and Group D: the rats receiving pioglitazone. Interleukin (IL) -6, IL-1β, tumor necrosis factor alpha (TNF-α), nitric-oxide (NO) in serum blood and superoxide dismutase (SOD), catalase (CAT) malondialdehyde, (MDA) and myeloperoxidase (MPO) in liver and spleen homogenates were measured. The amount of antioxidant enzymes in the spleen and liver, and finally cell viability and rats' survival were investigated. The measurement of blood serum nitric-oxide and survival of all groups of rats were also performed. The results indicated that both drugs could cause a decrease in IL-1β, IL-6, TNF-α and NO in rat blood serum and MDA and MPO in the liver and spleen homogenates, however, a significant increase in SOD and CAT in the liver and spleen homogenates in rats that received ANO and pioglitazone was observed compared to rats with CLP group. Cell viability and rats' survival were significantly improved in rats that received ANO and pioglitazone compared to rats with CLP group. Adenosine N1-oxide showed stronger and more effective effects.
Collapse
Affiliation(s)
- Yaser Jafari-Khataylou
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran;
| | - Siamak Kazemi-Darabadi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran;
| | - Somayeh Ahmadi Afshar
- PhD Candidate, Department of Microbiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| |
Collapse
|
16
|
Hasankhani A, Bahrami A, Tavakoli-Far B, Iranshahi S, Ghaemi F, Akbarizadeh MR, Amin AH, Abedi Kiasari B, Mohammadzadeh Shabestari A. The role of peroxisome proliferator-activated receptors in the modulation of hyperinflammation induced by SARS-CoV-2 infection: A perspective for COVID-19 therapy. Front Immunol 2023; 14:1127358. [PMID: 36875108 PMCID: PMC9981974 DOI: 10.3389/fimmu.2023.1127358] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/08/2023] [Indexed: 02/19/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a severe respiratory disease caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that affects the lower and upper respiratory tract in humans. SARS-CoV-2 infection is associated with the induction of a cascade of uncontrolled inflammatory responses in the host, ultimately leading to hyperinflammation or cytokine storm. Indeed, cytokine storm is a hallmark of SARS-CoV-2 immunopathogenesis, directly related to the severity of the disease and mortality in COVID-19 patients. Considering the lack of any definitive treatment for COVID-19, targeting key inflammatory factors to regulate the inflammatory response in COVID-19 patients could be a fundamental step to developing effective therapeutic strategies against SARS-CoV-2 infection. Currently, in addition to well-defined metabolic actions, especially lipid metabolism and glucose utilization, there is growing evidence of a central role of the ligand-dependent nuclear receptors and peroxisome proliferator-activated receptors (PPARs) including PPARα, PPARβ/δ, and PPARγ in the control of inflammatory signals in various human inflammatory diseases. This makes them attractive targets for developing therapeutic approaches to control/suppress the hyperinflammatory response in patients with severe COVID-19. In this review, we (1) investigate the anti-inflammatory mechanisms mediated by PPARs and their ligands during SARS-CoV-2 infection, and (2) on the basis of the recent literature, highlight the importance of PPAR subtypes for the development of promising therapeutic approaches against the cytokine storm in severe COVID-19 patients.
Collapse
Affiliation(s)
- Aliakbar Hasankhani
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Abolfazl Bahrami
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
- Faculty of Agricultural Sciences and Engineering, University of Tehran, Karaj, Iran
| | - Bahareh Tavakoli-Far
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Department of Physiology and Pharmacology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Setare Iranshahi
- School of Pharmacy, Shahid Beheshty University of Medical Sciences, Tehran, Iran
| | - Farnaz Ghaemi
- Department of Biochemistry, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Majid Reza Akbarizadeh
- Department of Pediatric, School of Medicine, Amir al momenin Hospital, Zabol University of Medical Sciences, Zabol, Iran
| | - Ali H. Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Bahman Abedi Kiasari
- Virology Department, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Alireza Mohammadzadeh Shabestari
- Department of Dental Surgery, Mashhad University of Medical Sciences, Mashhad, Iran
- Khorasan Covid-19 Scientific Committee, Mashhad, Iran
| |
Collapse
|
17
|
Ex Vivo Evaluation of Glutamine Treatment in Sepsis and Trauma in a Human Peripheral Blood Mononuclear Cells Model. Nutrients 2023; 15:nu15010252. [PMID: 36615909 PMCID: PMC9824313 DOI: 10.3390/nu15010252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/21/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023] Open
Abstract
We aimed to assess the lipopolysaccharide (LPS), or heat shock (HS) induction, and glutamine-modulating effects on heat shock protein-90α (HSP90α) and cytokines in an ex vivo model using peripheral blood mononuclear cells (PBMCs). The PBMCs of patients with septic shock, trauma-related systemic inflammatory response syndrome (SIRS), and healthy subjects were incubated with 1 μg/mL LPS at 43 °C (HS). Glutamine 10 mM was added 1 hour before or after induction or not at all. We measured mRNA HSP90α, monocyte (m) and lymphocyte (l) HSP90α proteins, interleukin (IL)-1b, -6, -8, -10, tumor necrosis factor-α (TNF-α), and monocyte chemoattractant protein-1 (MCP-1) supernatant levels. Heat shock increased the HSP90α mRNA and mHSP90α in all groups (10-fold in sepsis, p < 0.001 and p = 0.047, respectively). LPS induced the mHSP90α and lHSP90α in healthy (p < 0.001) and mHSP90α in SIRS (p = 0.004) but not in sepsis. LPS induced the cytokines at 24 and 48 h in all groups, especially in trauma (p < 0.001); HS only induced the IL-8 in healthy (p = 0.003) and septic subjects (p = 0.05). Glutamine at 10 mM before or after stimulation did not alter any induction effect of LPS or HS on HSP90α mRNA and mHSP90α protein in sepsis. In SIRS, glutamine before LPS decreased the mHSP90α but increased it when given after HS (p = 0.018). Before or after LPS (p = 0.049) and before HS (p = 0.018), glutamine decreased the lHSP90α expression in sepsis but increased it in SIRS when given after HS (p = 0.003). Regarding cytokines, glutamine enhanced the LPS-induced MCP-1 at 48 h in healthy (p = 0.011), SIRS (p < 0.001), and sepsis (p = 0.006). In conclusion, glutamine at 10 mM, before or after LPS and HS, modulates mHSP90α and lHSP90α in sepsis and SIRS differently and unpredictably. Although it does not alter the stimulation effect on interleukins, glutamine enhances the LPS induction effect on supernatant MCP-1 in all groups. Future research should seek to elucidate better the impact of glutamine and temperature modulation on HSP90α and MCP-1 pathways in sepsis and trauma.
Collapse
|
18
|
Interleukin-33 inhibits glucose uptake in human adipocytes and its expression in adipose tissue is elevated in insulin resistance and type 2 diabetes. Cytokine 2023; 161:156080. [PMID: 36368230 DOI: 10.1016/j.cyto.2022.156080] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/05/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Interleukin-33 (IL-33) is associated with obesity-related inflammation. We aim to investigate IL-33 expression in subcutaneous adipose tissue (SAT) in type 2 diabetes (T2D) subjects and its effects on human adipocyte glucose uptake. METHODS Expression of IL-33 was analysed in SAT from cohort studies including subjects with and without obesity and T2D and correlated with insulin resistance and obesity markers. Magnetic resonance imaging (MRI) of tissue fat volumes was performed. We investigated the effects of IL-33 treatment on ex vivo adipocyte glucose uptake. RESULTS T2D subjects had higher IL-33 gene and protein expression in SAT than the control subjects. IL-33 mRNA expression was positively correlated with markers of dysglycemia (e.g. HbA1c), insulin resistance (e.g. HOMA-IR) and adiposity (BMI, visceral adipose tissue volume, liver and pancreas fat %). In multiple linear regression analyses, insulin resistance and T2D status were the strongest predictors of IL-33, independent of BMI. IL-33 mRNA expression was negatively correlated with expression of genes regulating adipocyte glucose uptake, lipid storage, and adipogenesis (e.g.glucose transporter 1 and 4 (GLUT1/4), fatty acid binding protein 4 (FABP4), and PPARG). Additionally, incubation of SAT with IL-33 reduced adipocyte glucose uptake and GLUT4 gene and protein expression. CONCLUSIONS Our findings suggest that T2D subjects have higher IL-33 gene and protein expressionin SATthan control subjects, which is associated with insulin resistance and reduced gene expression of lipid storage and adipogenesis markers. IL-33 may reduce adipocyte glucose uptake. This opens up a potential pharmacological route for reversing insulin resistance in T2D and prediabetes.
Collapse
|
19
|
Fitton R, Sweetman J, Heseltine-Carp W, van der Feltz-Cornelis C. Anti-inflammatory medications for the treatment of mental disorders: A scoping review. Brain Behav Immun Health 2022; 26:100518. [PMID: 36217374 PMCID: PMC9547233 DOI: 10.1016/j.bbih.2022.100518] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/15/2022] [Accepted: 09/18/2022] [Indexed: 11/25/2022] Open
Abstract
This scoping review assessed the effect of anti-inflammatory medications in mental disorders. A search in Medline and the Cochrane database focusing on randomised controlled trials and systematic reviews identified 53 primary research articles, conducted in major depression, bipolar disorder, schizophrenia and somatic symptom disorders and related disorders (SSRD). The findings suggest that there is scope to consider the use of anti-inflammatory agents in mental disorders, however, not as a one-size-fits-all solution. Treatment could be especially helpful in subgroups with evidence of baseline inflammation. Anti-inflammatory medications that seem mostly effective in bipolar disorder or major depressive disorder, such as Celecoxib, Pioglitazone and statins, may differ from the ones with indications of effectiveness in schizophrenia, such as Minocycline and Aspirin. This might suggest a different underlying mechanism for treatment success in those two main illness groups. Further studies with larger sample sizes are needed that take levels of inflammation markers into account.
Collapse
Affiliation(s)
- Rebecca Fitton
- Kings College London, London, United Kingdom
- Tees Esk and Wear Valley NHS Foundation Trust, Darlington, United Kingdom
- Leeds and York Partnership NHS Foundation Trust, Leeds, United Kingdom
| | | | - William Heseltine-Carp
- Hull York Medical School (HYMS), University of York, York, United Kingdom
- Hull University Teaching Hospitals NHS Trust, United Kingdom
| | - Christina van der Feltz-Cornelis
- Tees Esk and Wear Valley NHS Foundation Trust, Darlington, United Kingdom
- Dept of Health Sciences, University of York, York, United Kingdom
- Hull York Medical School (HYMS), University of York, York, United Kingdom
- Institute of Health Informatics, University College London, London, United Kingdom
| |
Collapse
|
20
|
Miyashita D, Inoue R, Tsuno T, Okuyama T, Kyohara M, Nakahashi-Oda C, Nishiyama K, Fukushima S, Inada Y, Togashi Y, Shibuya A, Terauchi Y, Shirakawa J. Protective effects of S100A8 on sepsis mortality: Links to sepsis risk in obesity and diabetes. iScience 2022; 25:105662. [PMID: 36505926 PMCID: PMC9732389 DOI: 10.1016/j.isci.2022.105662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/23/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Obesity and diabetes are independent risk factors for death during sepsis. S100A8, an alarmin, is related to inflammation, obesity, and diabetes. Here, we examine the role of S100A8 in sepsis of obesity and diabetes models. Injection of S100A8 prolongs the survival of septic mice induced by lethal endotoxemia, Escherichia coli injection, or cecal ligation and puncture. S100A8 decrease the LPS-induced expression of proinflammatory cytokines in peritoneal macrophages by inhibiting TLR4-mediated signals in an autocrine manner. db/db, ob/ob, and western diet-fed mice demonstrate reduced upregulation of S100A8 induced by LPS treatment in both serum and peritoneal cells. These mice also show shorter survival after LPS injection, and S100A8 supplementation prolonged the survival. While myelomonocytic cells-specific S100A8-deficient mice (Lyz2 cre :S100A8 floxed/floxed ) exhibit shorter survival after LPS treatment, S100A8 supplementation prolonged the survival. Thus, myelomonocytic cell-derived S100A8 is crucial for protection from sepsis, and S100A8 supplementation improves sepsis, particularly in mice with obesity and diabetes.
Collapse
Affiliation(s)
- Daisuke Miyashita
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Ryota Inoue
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Takahiro Tsuno
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Tomoko Okuyama
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Mayu Kyohara
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Chigusa Nakahashi-Oda
- Department of Immunology, Faculty of Medicine, and R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Japan
| | - Kuniyuki Nishiyama
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Setsuko Fukushima
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
| | - Yutaro Inada
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Yu Togashi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, and R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Jun Shirakawa
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
- Corresponding author
| |
Collapse
|
21
|
Miyazaki H, Kinoshita M, Nakashima H, Nakamura S, Saitoh D. Pioglitazone Modifies Kupffer Cell Function and Protects against Escherichia coli-Induced Bacteremia in Burned Mice. Int J Mol Sci 2022; 23:12746. [PMID: 36361535 PMCID: PMC9657905 DOI: 10.3390/ijms232112746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/12/2022] [Accepted: 10/20/2022] [Indexed: 01/24/2023] Open
Abstract
Infectious complications and subsequent sepsis in severely burned patients lead to high morbidity and mortality in response to uncontrolled innate immune responses mediated by macrophages. Peroxisome proliferator-activated receptor gamma (PPARγ) has anti-inflammatory activity and acts as a master regulator of macrophage polarization. In this study, we investigated whether the administration of a PPARγ agonist could modulate the Kupffer cell phenotype and thereby ameliorate the dysregulated innate response during post-burn bacterial infection. C57BL/6 mice were subjected to severe burns and randomized to receive either the PPARγ agonist, pioglitazone, or the vehicle control five days after injury, followed by the subsequent analysis of hepatic macrophages. Survival from the bacterial infection was monitored for seven days. Pioglitazone protected burned mice against bacterial infection. A single treatment with pioglitazone significantly enhanced phagocytosis, phagosome acidification, bacterial clearance, and reduction in inflammatory mediators in Kupffer cells. In conclusion, PPARγ activation by pioglitazone prevents clinical deterioration due to post-burn bacterial infection and improves survival. Our findings suggest that pioglitazone may be an effective therapeutic candidate for post-burn infectious complications.
Collapse
Affiliation(s)
- Hiromi Miyazaki
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Saitama 359-8513, Japan
| | - Manabu Kinoshita
- Department of Immunology and Microbiology, National Defense Medical College, Saitama 359-8513, Japan
| | - Hiroyuki Nakashima
- Department of Immunology and Microbiology, National Defense Medical College, Saitama 359-8513, Japan
| | - Shingo Nakamura
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Saitama 359-8513, Japan
| | - Daizoh Saitoh
- Division of Traumatology, National Defense Medical College Research Institute, Saitama 359-8513, Japan
| |
Collapse
|
22
|
Kynurenine Pathway-An Underestimated Factor Modulating Innate Immunity in Sepsis-Induced Acute Kidney Injury? Cells 2022; 11:cells11162604. [PMID: 36010680 PMCID: PMC9406744 DOI: 10.3390/cells11162604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection, and it accounts for about half of the cases of acute kidney injury (AKI). Although sepsis is the most frequent cause of AKI in critically ill patients, its pathophysiological mechanisms are not well understood. Sepsis has the ability to modulate the function of cells belonging to the innate immune system. Increased activity of indoleamine 2,3-dioxygenase 1 (IDO1) and production of kynurenines are the major metabolic pathways utilized by innate immunity cells to maintain immunological tolerance. The activation of the kynurenine pathway (KP) plays a dual role in sepsis—in the early stage, the induction of IDO1 elicits strong proinflammatory effects that may lead to tissue damage and septic shock. Afterwards, depletion of tryptophan and production of kynurenines contribute to the development of immunosuppression that may cause the inability to overpower opportunistic infections. The presented review provides available data on the various interdependencies between elements of innate immunity and sepsis-induced AKI (SAKI) with particular emphasis on the immunomodulatory significance of KP in the above processes. We believe that KP activation may be one of the crucial, though underestimated, components of a deregulated host response to infection during SAKI.
Collapse
|
23
|
Neuroprotective and Anti-inflammatory Effects of Pioglitazone on Traumatic Brain Injury. Mediators Inflamm 2022; 2022:9860855. [PMID: 35757108 PMCID: PMC9232315 DOI: 10.1155/2022/9860855] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/06/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) is still a major cause of concern for public health, and out of all the trauma-related injuries, it makes the highest contribution to death and disability worldwide. Patients of TBI continue to suffer from brain injury through an intricate flow of primary and secondary injury events. However, when treatment is provided in a timely manner, there is a significant window of opportunity to avoid a few of the serious effects. Pioglitazone (PG), which has a neuroprotective impact and can decrease inflammation after TBI, activates peroxisome proliferator-activated receptor-gamma (PPARγ). The objective of the study is to examine the existing literature to assess the neuroprotective and anti-inflammatory impact of PG in TBI. It also discusses the part played by microglia and cytokines in TBI. According to the findings of this study, PG has the ability to enhance neurobehavior, decrease brain edema and neuronal injury following TBI. To achieve the protective impact of PG the following was required: (1) stimulating PPARγ; (2) decreasing oxidative stress; (3) decreasing nuclear factor kappa B (NF-κB), interleukin 6 (IL-6), interleukin-1β (IL-1β), cyclooxygenase-2 (COX-2), and C-C motif chemokine ligand 20 (CCL20) expression; (4) limiting the increase in the number of activated microglia; and (5) reducing mitochondrial dysfunction. The findings indicate that when PIG is used clinically, it may serve as a neuroprotective anti-inflammatory approach in TBI.
Collapse
|
24
|
Wang YF, Li JW, Wang DP, Jin K, Hui JJ, Xu HY. Anti-Hyperglycemic Agents in the Adjuvant Treatment of Sepsis: Improving Intestinal Barrier Function. Drug Des Devel Ther 2022; 16:1697-1711. [PMID: 35693534 PMCID: PMC9176233 DOI: 10.2147/dddt.s360348] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/28/2022] [Indexed: 12/19/2022] Open
Abstract
Intestinal barrier injury and hyperglycemia are common in patients with sepsis. Bacteria translocation and systemic inflammatory response caused by intestinal barrier injury play a significant role in sepsis occurrence and deterioration, while hyperglycemia is linked to adverse outcomes in sepsis. Previous studies have shown that hyperglycemia is an independent risk factor for intestinal barrier injury. Concurrently, increasing evidence has indicated that some anti-hyperglycemic agents not only improve intestinal barrier function but are also beneficial in managing sepsis-induced organ dysfunction. Therefore, we assume that these agents can block or reduce the severity of sepsis by improving intestinal barrier function. Accordingly, we explicated the connection between sepsis, intestinal barrier, and hyperglycemia, overviewed the evidence on improving intestinal barrier function and alleviating sepsis-induced organ dysfunction by anti-hyperglycemic agents (eg, metformin, peroxisome proliferators activated receptor-γ agonists, berberine, and curcumin), and summarized some common characteristics of these agents to provide a new perspective in the adjuvant treatment of sepsis.
Collapse
Affiliation(s)
- Yi-Feng Wang
- Department of Critical Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, People's Republic of China
| | - Jia-Wei Li
- Department of Critical Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, People's Republic of China
| | - Da-Peng Wang
- Department of Critical Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, People's Republic of China
| | - Ke Jin
- Department of Critical Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, People's Republic of China
| | - Jiao-Jie Hui
- Department of Critical Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, People's Republic of China
| | - Hong-Yang Xu
- Department of Critical Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, People's Republic of China
| |
Collapse
|
25
|
Naz S, Al Kury LT, Nadeem H, Shah FA, Ullah A, Paracha RZ, Imran M, Li S. Synthesis, In Silico and Pharmacological Evaluation of New Thiazolidine-4-Carboxylic Acid Derivatives Against Ethanol-Induced Neurodegeneration and Memory Impairment. J Inflamm Res 2022; 15:3643-3660. [PMID: 35783245 PMCID: PMC9241999 DOI: 10.2147/jir.s357082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 06/04/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Several studies revealed that alcohol utilization impairs memory in adults; however, the underlying mechanism is still unclear. The production of inflammatory markers and reactive oxygen species (ROS) plays a major role in neurodegeneration, which leads to memory impairment. Therefore, targeting neuroinflammation and oxidative distress could be a useful strategy for abrogating the hallmarks of ethanol-induced neurodegeneration. Moreover, several studies have demonstrated multiple biological activities of thiazolidine derivatives including neuroprotection. Methods In the current study, we synthesized ten (10) new thiazolidine-4-carboxylic acid derivatives (P1-P10), characterized their synthetic properties using proton nuclear magnetic resonance (1H-NMR) and carbon-13 NMR, and further investigated the neuroprotective potential of these compounds in an ethanol-induced neuroinflammation model. Results Our results suggested altered levels of antioxidant enzymes associated with an elevated level of tumor necrosis factor-alpha (TNF-α), nuclear factor-κB (p-NF-κB), pyrin domain-containing protein 3 (NLRP3), and cyclooxygenase-2 (COX-2) in ethanol-treated animals. Ethanol treatment also led to memory impairment in rats, as assessed by behavioral tests. To further support our notion, we performed molecular docking studies, and all synthetic compounds exhibited a good binding affinity with a fair bond formation with selected targets (NF-κB, TLR4, NLRP3, and COX-2). Discussion Overall, our results revealed that these derivatives may be beneficial in reducing neuroinflammation by acting on different stages of inflammation. Moreover, P8 and P9 treatment attenuated the neuroinflammation, oxidative stress, and memory impairment caused by ethanol.
Collapse
Affiliation(s)
- Shagufta Naz
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, People’s Republic of China
| | - Lina Tariq Al Kury
- College of Natural and Health Sciences, Zayed University, Abu Dhabi, 49153, United Arab Emirates
| | - Humaira Nadeem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
- Correspondence: Humaira Nadeem, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan, Tel +92 51-2891835, Fax +92 51-8350180, Email
| | - Fawad Ali Shah
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
| | - Aman Ullah
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
| | - Rehan Zafar Paracha
- Research Center for Modeling & Simulation (RCMS), National University of Sciences and Technology (NUST), Islamabad, 44000, Pakistan
| | - Muhammad Imran
- Department of Pharmacy, IQRA University, Islamabad, 44000, Pakistan
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, People’s Republic of China
- Shupeng Li, State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, People’s Republic of China, Email
| |
Collapse
|
26
|
Reducing neuroinflammation via therapeutic compounds and lifestyle to prevent or delay progression of Parkinson's disease. Ageing Res Rev 2022; 78:101618. [PMID: 35395416 DOI: 10.1016/j.arr.2022.101618] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/08/2022] [Accepted: 04/01/2022] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is the second most common age-associated neurodegenerative disorder and is characterised by progressive loss of dopamine neurons in the substantia nigra. Peripheral immune cell infiltration and activation of microglia and astrocytes are observed in PD, a process called neuroinflammation. Neuroinflammation is a fundamental response to protect the brain but, when chronic, it triggers neuronal damage. In the last decade, central and peripheral inflammation were suggested to occur at the prodromal stage of PD, sustained throughout disease progression, and may play a significant role in the pathology. Understanding the pathological mechanisms of PD has been a high priority in research, primarily to find effective treatments once symptoms are present. Evidence indicates that early life exposure to neuroinflammation as a consequence of life events, environmental or behaviour factors such as exposure to infections, pollution or a high fat diet increase the risk of developing PD. Many studies show healthy habits and products that decrease neuroinflammation also reduce the risk of PD. Here, we aim to stimulate discussion about the role of neuroinflammation in PD onset and progression. We highlight that reducing neuroinflammation throughout the lifespan is critical for preventing idiopathic PD, and present epidemiological studies that detail risk and protective factors. It is possible that introducing lifestyle changes that reduce neuroinflammation at the time of PD diagnosis may slow symptom progression. Finally, we discuss compounds and therapeutics to treat the neuroinflammation associated with PD.
Collapse
|
27
|
Lin X, Meng X, Song Z, Lin J. Peroxisome proliferator-activator receptor γ and psoriasis, molecular and cellular biochemistry. Mol Cell Biochem 2022; 477:1905-1920. [PMID: 35348980 DOI: 10.1007/s11010-022-04417-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/16/2022] [Indexed: 10/18/2022]
Abstract
The pathophysiology of psoriasis is complex and has not been completely elucidated. Better understanding of the pathogenesis may contribute to further improvement of our therapeutic strategies controlling psoriasis. Emerging evidence points to a causative relationship between altered activity of peroxisome proliferator-activated receptor γ (PPARγ) and psoriasis. The present review focuses on deeper understanding of the possible role of PPARγ in the pathogenesis of psoriasis and the potential of PPARγ agonist to improve the treatment of psoriasis. PPARγ is decreased in psoriasis. PPARγ possibly has effects on the multiple aspects of the pathogenesis of psoriasis, including abnormal lipid metabolism, insulin resistance, immune cells, pro-inflammatory cytokines, keratinocytes, angiogenesis, oxidative stress, microRNAs and nuclear factor kappa B. As defective activation of PPARγ is involved in psoriasis development, PPARγ agonists may be promising agents for treatment of psoriasis. Pioglitazone appears an effective and safe option in the treatment of patients with psoriasis, but there are still concerns about its potential side effects. Research effort has recently been undertaken to explore the PPARγ-activating potential of natural products. Among them some have been studied clinically or preclinically for treatment of psoriasis with promising results.
Collapse
Affiliation(s)
- Xiran Lin
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Lu, Dalian, 116011, China.
| | - Xianmin Meng
- Department of Pathology and Laboratory Medicine, Axia Women's Health, 450 Cresson BLVD, Oaks, PA, 19456, USA
| | - Zhiqi Song
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Lu, Dalian, 116011, China
| | - Jingrong Lin
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Lu, Dalian, 116011, China
| |
Collapse
|
28
|
Zeng H, Dumitrescu AV, Wadkins D, Elwood BW, Gramlich OW, Kuehn MH. Systemic Treatment with Pioglitazone Reverses Vision Loss in Preclinical Glaucoma Models. Biomolecules 2022; 12:281. [PMID: 35204782 PMCID: PMC8961625 DOI: 10.3390/biom12020281] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
Neuroinflammation significantly contributes to the pathophysiology of several neurodegenerative diseases. This is also the case in glaucoma and may be a reason why many patients suffer from progressive vision loss despite maximal reduction in intraocular pressure. Pioglitazone is an agonist of the peroxisome proliferator-activated receptor gamma (PPARγ) whose pleiotrophic activities include modulation of cellular energy metabolism and reduction in inflammation. In this study we employed the DBA2/J mouse model of glaucoma with chronically elevated intraocular pressure to investigate whether oral low-dose pioglitazone treatment preserves retinal ganglion cell (RGC) survival. We then used an inducible glaucoma model in C57BL/6J mice to determine visual function, pattern electroretinographs, and tracking of optokinetic reflex. Our findings demonstrate that pioglitazone treatment does significantly protect RGCs and prevents axonal degeneration in the glaucomatous retina. Furthermore, treatment preserves and partially reverses vision loss in spite of continuously elevated intraocular pressure. These data suggest that pioglitazone may provide treatment benefits for those glaucoma patients experiencing continued vision loss.
Collapse
Affiliation(s)
- Huilan Zeng
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha 410011, China;
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA 52242, USA; (A.V.D.); (D.W.); (B.W.E.); (O.W.G.)
- Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA
- Human Clinical Research Center of Ophthalmic Disease, Changsha 410011, China
| | - Alina V. Dumitrescu
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA 52242, USA; (A.V.D.); (D.W.); (B.W.E.); (O.W.G.)
| | - David Wadkins
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA 52242, USA; (A.V.D.); (D.W.); (B.W.E.); (O.W.G.)
- Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA
| | - Benjamin W. Elwood
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA 52242, USA; (A.V.D.); (D.W.); (B.W.E.); (O.W.G.)
- Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA
| | - Oliver W. Gramlich
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA 52242, USA; (A.V.D.); (D.W.); (B.W.E.); (O.W.G.)
- Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA
| | - Markus H. Kuehn
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA 52242, USA; (A.V.D.); (D.W.); (B.W.E.); (O.W.G.)
- Iowa City VA Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA
| |
Collapse
|
29
|
Liu SY, Huang CC, Huang SF, Liao TL, Kuo NR, Yang YY, Li TH, Liu CW, Hou MC, Lin HC. Pioglitazone Ameliorates Acute Endotoxemia-Induced Acute on Chronic Renal Dysfunction in Cirrhotic Ascitic Rats. Cells 2021; 10:3044. [PMID: 34831270 PMCID: PMC8616474 DOI: 10.3390/cells10113044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 01/23/2023] Open
Abstract
Endotoxemia-activated tumor necrosis factor (TNFα)/nuclear factor kappa B (NFκB) signals result in acute on chronic inflammation-driven renal dysfunction in advanced cirrhosis. Systemic activation of peroxisome proliferator-activated receptor gamma (PPARγ) with pioglitazone can suppress inflammation-related splanchnic and pulmonary dysfunction in cirrhosis. This study explored the mechanism and effects of pioglitazone treatment on the abovementioned renal dysfunction in cirrhotic rats. Cirrhotic ascitic rats were induced with renal dysfunction by bile duct ligation (BDL). Then, 2 weeks of pioglitazone treatment (Pio, PPAR gamma agonist, 12 mg/kg/day, using the azert osmotic pump) was administered from the 6th week after BDL. Additionally, acute lipopolysaccharide (LPS, Escherichia coli 0111:B4; Sigma, 0.1 mg/kg b.w, i.p. dissolved in NaCl 0.9%) was used to induce acute renal dysfunction. Subsequently, various circulating, renal arterial and renal tissue pathogenic markers were measured. Cirrhotic BDL rats are characterized by decreased mean arterial pressure, increased cardiac output and portal venous pressure, reduced renal arterial blood flow (RABF), increased renal vascular resistance (RVR), increased relative renal weight/hydroxyproline, downregulated renal PPARγ expression, upregulated renal inflammatory markers (TNFα, NFκB, IL-6, MCP-1), increased adhesion molecules (VCAM-1 and ICAM-1), increased renal macrophages (M1, CD68), and progressive renal dysfunction (increasing serum and urinary levels of renal injury markers (lipocalin-2 and IL-18)). In particular, acute LPS administration induces acute on chronic renal dysfunction (increasing serum BUN/creatinine, increasing RVR and decreasing RABF) by increased TNFα-NFκB-mediated renal inflammatory markers as well as renal M1 macrophage infiltration. In comparison with the BDL+LPS group, chronic pioglitazone pre-treatment prevented LPS-induced renal pathogenic changes in the BDL-Pio+LPS group. Activation of systemic, renal vessel and renal tissue levels of PPARγ by chronic pioglitazone treatment has beneficial effects on the endotoxemia-related TNFα/NFκB-mediated acute and chronic renal inflammation in cirrhosis. This study revealed that normalization of renal and renal arterial levels of PPARγ effectively prevented LPS-induced acute and chronic renal dysfunction in cirrhotic ascitic rats.
Collapse
Affiliation(s)
- Szu-Yu Liu
- Department of Medical Education, Medical Innovation and Research Office (MIRO), Taipei Veterans General Hospital, Taipei 11217, Taiwan; (S.-Y.L.); (C.-C.H.); (N.-R.K.)
- Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (S.-F.H.); (M.-C.H.)
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan; (T.-L.L.); (T.-H.L.); (C.-W.L.)
| | - Chia-Chang Huang
- Department of Medical Education, Medical Innovation and Research Office (MIRO), Taipei Veterans General Hospital, Taipei 11217, Taiwan; (S.-Y.L.); (C.-C.H.); (N.-R.K.)
- Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (S.-F.H.); (M.-C.H.)
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan; (T.-L.L.); (T.-H.L.); (C.-W.L.)
| | - Shiang-Fen Huang
- Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (S.-F.H.); (M.-C.H.)
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan; (T.-L.L.); (T.-H.L.); (C.-W.L.)
| | - Tsai-Ling Liao
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan; (T.-L.L.); (T.-H.L.); (C.-W.L.)
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 11217, Taiwan
| | - Nai-Rong Kuo
- Department of Medical Education, Medical Innovation and Research Office (MIRO), Taipei Veterans General Hospital, Taipei 11217, Taiwan; (S.-Y.L.); (C.-C.H.); (N.-R.K.)
- Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (S.-F.H.); (M.-C.H.)
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan; (T.-L.L.); (T.-H.L.); (C.-W.L.)
| | - Ying-Ying Yang
- Department of Medical Education, Medical Innovation and Research Office (MIRO), Taipei Veterans General Hospital, Taipei 11217, Taiwan; (S.-Y.L.); (C.-C.H.); (N.-R.K.)
- Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (S.-F.H.); (M.-C.H.)
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan; (T.-L.L.); (T.-H.L.); (C.-W.L.)
| | - Tzu-Hao Li
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan; (T.-L.L.); (T.-H.L.); (C.-W.L.)
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Foundation, Taipei 11217, Taiwan
| | - Chih-Wei Liu
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan; (T.-L.L.); (T.-H.L.); (C.-W.L.)
| | - Ming-Chih Hou
- Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (S.-F.H.); (M.-C.H.)
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan; (T.-L.L.); (T.-H.L.); (C.-W.L.)
| | - Han-Chieh Lin
- Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (S.-F.H.); (M.-C.H.)
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan; (T.-L.L.); (T.-H.L.); (C.-W.L.)
| |
Collapse
|
30
|
Brayner Cavalcanti M, Bezerra Leite LL, Manoel de Queiroz D, de Salazar E Fernandes T, Mendes de Oliveira WL, Pereira MC, da Rocha Pitta MG, de Melo Rêgo MJB, Dos Santos Júnior JA, Herrero Fernández Z, Rodrigues Cravo Teixeira D, Galdino da Rocha Pitta M, da Rocha Pitta I, César Dantas S, Cavalcante Veras R, Almeida de Medeiros I, Borges da Silva E. Evaluation of the action of LPSF/GQ-16 on cytokines and PPAR-γ gene expression after in vitro irradiation of peripheral blood mononuclear cells. Int J Radiat Biol 2021; 97:1649-1656. [PMID: 34586957 DOI: 10.1080/09553002.2021.1987556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE Patients submitted to radiotherapy (RT) may present in their healthy tissues surrounding the treated tumor, some typical acute inflammatory reactions induced by ionizing radiation (IR). The manifestation of inflammatory processes is a result of exacerbation of the immune system, as a response to radiation exposure, and this can be a limiting factor for RT protocols. To counteract this, some thiazolidinediones, such as LPSF/GQ-16, may be useful for modulating the patient's radioinduced inflammatory response in normal tissues. In this context, the present work aims to evaluate the activity of LPSF/GQ-16 on the levels of cytokines and the expression of the gene PPARγ in mononuclear cells irradiated in vitro, to analyze the immunomodulatory activity of the molecule and its action on radiomitigation. MATERIALS AND METHODS For this, blood samples from eight donors were collected and irradiated with 2 Gy, then the PBMC (peripheral blood mononuclear cells) were cultured and treated with LPSF/GQ-16. The levels of cytokines TNF-α, IFN-γ, IL-2 and IL-4 were quantified by CBA, while the genes of TNF-α, IFN-γ and PPARγ were analyzed by RT-PCR. RESULTS LPSF/GQ-16 significantly reduced the expression of proinflammatory cytokines (IFN-γ and TNF-α) in irradiated and nonirradiated groups. There was no significant reduction of anti-inflammatory cytokines (IL-2 and IL-4) by LPSF/GQ-16. The mRNA expression of PPAR-γ, IFN-γ and TNF-α in the presence of LPSF/GQ-16 was higher in the nonirradiated sample. CONCLUSION LPSF/GQ-16 showed effective activity after irradiation, with an important immunomodulatory activity in irradiated PBMCs.
Collapse
Affiliation(s)
- Mariana Brayner Cavalcanti
- Grupo de Radioecologia, Departamento de Energia Nuclear, Universidade Federal de Pernambuco, Recife, Brazil
| | - Lídia Lúcia Bezerra Leite
- Grupo de Estudos em Radioproteção e Radioecologia (GERAR), Departamento de Energia Nuclear, Universidade Federal de Pernambuco, Recife, Brazil
| | - Diogo Manoel de Queiroz
- Grupo de Estudos em Radioproteção e Radioecologia (GERAR), Departamento de Energia Nuclear, Universidade Federal de Pernambuco, Recife, Brazil
| | - Thiago de Salazar E Fernandes
- Grupo de Radioecologia, Departamento de Energia Nuclear, Universidade Federal de Pernambuco, Recife, Brazil.,Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Brazil
| | - Wagner Luís Mendes de Oliveira
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas (LINAT), Núcleo de Pesquisas em Inovação Terapêutica Suely Galdino (NUPIT SG), Universidade Federal de Pernambuco, Recife, Brazil
| | - Michelly Cristiny Pereira
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas (LINAT), Núcleo de Pesquisas em Inovação Terapêutica Suely Galdino (NUPIT SG), Universidade Federal de Pernambuco, Recife, Brazil
| | - Maíra Galdino da Rocha Pitta
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas (LINAT), Núcleo de Pesquisas em Inovação Terapêutica Suely Galdino (NUPIT SG), Universidade Federal de Pernambuco, Recife, Brazil
| | - Moacyr Jesus Barreto de Melo Rêgo
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas (LINAT), Núcleo de Pesquisas em Inovação Terapêutica Suely Galdino (NUPIT SG), Universidade Federal de Pernambuco, Recife, Brazil
| | | | - Zahily Herrero Fernández
- Grupo de Radioecologia, Departamento de Energia Nuclear, Universidade Federal de Pernambuco, Recife, Brazil
| | - Diego Rodrigues Cravo Teixeira
- Laboratório de Planejamento e Síntese de Fármacos, Núcleo de Pesquisas em Inovação Terapêutica Suely Galdino (NUPIT SG), Universidade Federal de Pernambuco, Recife, Brazil
| | - Marina Galdino da Rocha Pitta
- Laboratório de Planejamento e Síntese de Fármacos, Núcleo de Pesquisas em Inovação Terapêutica Suely Galdino (NUPIT SG), Universidade Federal de Pernambuco, Recife, Brazil
| | - Ivan da Rocha Pitta
- Laboratório de Planejamento e Síntese de Fármacos, Núcleo de Pesquisas em Inovação Terapêutica Suely Galdino (NUPIT SG), Universidade Federal de Pernambuco, Recife, Brazil
| | - Samuel César Dantas
- Centro de Radioterapia, Instituto Materno Infantil prof. Antônio Figueira, Recife, Brazil
| | - Robson Cavalcante Veras
- Instituto de Pesquisa em Fármacos e Medicamentos (IPeFarM/UFPB), Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde - Campus I, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Isac Almeida de Medeiros
- Instituto de Pesquisa em Fármacos e Medicamentos (IPeFarM/UFPB), Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde - Campus I, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Edvane Borges da Silva
- Grupo de Estudos em Radioproteção e Radioecologia (GERAR), Departamento de Energia Nuclear, Universidade Federal de Pernambuco, Recife, Brazil.,Centro Acadêmico de Vitória (CAV), Universidade Federal de Pernambuco, Recife, Brazil
| |
Collapse
|
31
|
Li C, Davis X, Lahni P, Stuck J, Williamson L, Kaplan J. Obesity protects against sepsis-induced and norepinephrine-induced white adipose tissue browning. Am J Physiol Endocrinol Metab 2021; 321:E433-E442. [PMID: 34370596 PMCID: PMC8461795 DOI: 10.1152/ajpendo.00380.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 11/22/2022]
Abstract
Sepsis is a dysregulated systemic response to infection and can lead to organ damage and death. Obesity is a significant problem worldwide and affects outcomes from sepsis. Our laboratory demonstrated that white adipose tissue (WAT) undergoes browning during sepsis, a process whereby WAT adopts a brown adipose tissue phenotype. However, this browning process was not observed in obese mice during sepsis. White adipose tissue browning is detrimental in patients with burn injury and cancer. We hypothesize that norepinephrine (NE) induces WAT browning in nonobese mice but not in obese mice similarly to sepsis-induced WAT browning. Six-week-old C57BL/6 male mice were randomized to a high-fat diet or normal diet. After 6-7 wk of feeding, polymicrobial sepsis was induced by cecal ligation and puncture (CLP). Norepinephrine was administered intraperitoneally via osmotic minipumps for 18 h or 72 h (no CLP) at which time tissue and plasma were harvested. Controls were mice that underwent CLP (no NE) with 18-h harvest. A separate group of mice underwent pretreatment with NE or vehicle infusion for 72 h, CLP was performed, and at 18 h had tissue and plasma harvested. Sepsis resulted in significant weight loss in both nonobese and obese mice. NE treatment alone caused weight loss in obese mice. Septic nonobese mice had higher uncoupling protein-1 (UCP1) expression compared with control and obese septic mice. NE treatment increased UCP1 expression in nonobese, but not obese mice. NE-treated obese septic mice had lower lung myeloperoxidase (MPO) activity, alanine aminotransferase (ALT), aspartate aminotransferase (AST), TNFα, and IL-6 levels compared with NE-treated nonobese septic mice. Obesity protects mice from septic-induced and NE-induced WAT browning.NEW & NOTEWORTHY White adipose tissue browning is detrimental in patients with burn injury and cancer. WAT browning occurs in nonobese mice and can be induced by β receptor norepinephrine infusion, but obese mice are resistant to sepsis-induced and norepinephrine-induced WAT browning. We propose that the lack of WAT browning and unchanged inflammatory cytokine response may contribute to the protection of obese mice from sepsis.
Collapse
Affiliation(s)
- Cheryl Li
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Xenia Davis
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Patrick Lahni
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joanna Stuck
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Lauren Williamson
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jennifer Kaplan
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
32
|
Victor S, Chew A, Falconer S. Pro12Ala polymorphism of peroxisome proliferator activated receptor gamma 2 may be associated with adverse neurodevelopment in European preterm babies. Brain Behav 2021; 11:e2256. [PMID: 34152086 PMCID: PMC8413715 DOI: 10.1002/brb3.2256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/28/2021] [Accepted: 06/06/2021] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Prematurity is the leading cause of death and disability in children under 5 years of age. Understanding the molecular mechanisms of the biological processes involved in preterm brain injury may help develop novel neuroprotective treatment strategies. A growing body of evidence suggest that peroxisome proliferator-activated receptor gamma (PPARγ) signaling is associated with inhibited brain development in preterm babies. The Ala allele of the Pro12Ala polymorphism of PPARγ2 decreases receptor binding affinity and consequently induces a reduction of PPARγ signaling. METHODS In this study, we carried out a preliminary analysis of existing datasets to test the hypothesis that reduced transactivation capacity of PPARγ in the presence of the Ala variant of PPARγ2 may be associated with adverse neurodevelopment in preterm babies. The association between PPAR-γ2 Pro12Ala polymorphism and neurodevelopment at 18-24 months of age was assessed in two groups of European infants, 155 born before 33 weeks' gestation and 180 born later than 36 weeks' gestation using a linear regression model. The Bayley Scales of Infant and Toddler Development-3rd edition was administered to assess neurodevelopment at 18-24 months of age. RESULTS We observed the Ala allele of the Pro12Ala polymorphism in 25% preterm infants and 20% term infants. The Ala allele of PPARγ2 was significantly associated with adverse cognitive (p = .019), language (p = .03), and motor development (p = 0.036) at 18-24 months of age after taking into consideration the duration of ventilation, gender, and index of multiple deprivation scores, but without correction for potential shared ancestry. There was no association between the PPAR-γ2 Pro12Ala polymorphism and neurodevelopment in term infants. CONCLUSIONS These preliminary data suggest that PPARγ signaling in the presence of the Ala variant of PPARγ2 may be associated with adverse neurodevelopment in preterm infants suggesting that further studies are warranted.
Collapse
Affiliation(s)
- Suresh Victor
- Department of Perinatal Imaging and HealthCentre for the Developing BrainSchool of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
| | - Andrew Chew
- Department of Perinatal Imaging and HealthCentre for the Developing BrainSchool of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
| | - Shona Falconer
- Department of Perinatal Imaging and HealthCentre for the Developing BrainSchool of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
| |
Collapse
|
33
|
Type 2 Diabetic Sepsis Patients Have a Lower Mortality Rate in Pioglitazone Use: A Nationwide 15-Year Propensity Score Matching Observational Study in Taiwan. Emerg Med Int 2021; 2021:4916777. [PMID: 34394992 PMCID: PMC8363455 DOI: 10.1155/2021/4916777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/06/2021] [Indexed: 12/18/2022] Open
Abstract
Background Pioglitazone use via the PPARγ agonist in sepsis patients is inconclusive. It was based on a great number of animal studies. However, except for information from animal studies, there are merely any data of human studies for reference. Methods This study was conducted by a unique database including 1.6 million diabetic patients. From 1999 to 2013, a total of 145,327 type 2 diabetic patients, first admitted for sepsis, were enrolled. Propensity score matching was conducted in a 1 : 5 ratio between pioglitazone users and nonusers. Multivariate logistic regression was conducted to evaluate the adjusted odds ratios (aORs) of hospital mortality in pioglitazone users. Further stratification analysis was done and Kaplan–Meier plot was used. Results A total of 9,310 sepsis pioglitazone users (defined as “ever” use of pioglitazone in any dose within 3 months prior to the first admission for sepsis) and 46,550 matched nonusers were retrieved, respectively. In the multivariate logistic regression model, the cohort of pioglitazone users (9,310) had a decreased aOR of 0.95 (95% CI, 0.89–1.02) of sepsis mortality. Further stratification analysis demonstrated that “chronic pioglitazone users” (defined as “at least” 4-week drug use within 3 months) (3,399) were more associated with significant aOR of 0.80 (95% CI, 0.72–0.89) in reducing sepsis mortality. Conclusions This first human cohort study demonstrated the potential protective effect of chronic pioglitazone use in type 2 diabetic sepsis patients.
Collapse
|
34
|
Jalilvand N, Hosseini M, Beheshti F, Ebrahimzadeh-Bideskan A. Protective effect of PPARγ agonist pioglitazone, on testicular tissue and sperm parameters in hypothyroid rats. TOXIN REV 2021; 40:267-276. [DOI: 10.1080/15569543.2018.1564775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 12/28/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Narjes Jalilvand
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farimah Beheshti
- Department of Basic Science and Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Alireza Ebrahimzadeh-Bideskan
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Microanatomy Research Center Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
35
|
Saunders AM, Burns DK, Gottschalk WK. Reassessment of Pioglitazone for Alzheimer's Disease. Front Neurosci 2021; 15:666958. [PMID: 34220427 PMCID: PMC8243371 DOI: 10.3389/fnins.2021.666958] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease is a quintessential 'unmet medical need', accounting for ∼65% of progressive cognitive impairment among the elderly, and 700,000 deaths in the United States in 2020. In 2019, the cost of caring for Alzheimer's sufferers was $244B, not including the emotional and physical toll on caregivers. In spite of this dismal reality, no treatments are available that reduce the risk of developing AD or that offer prolonged mitiagation of its most devestating symptoms. This review summarizes key aspects of the biology and genetics of Alzheimer's disease, and we describe how pioglitazone improves many of the patholophysiological determinants of AD. We also summarize the results of pre-clinical experiments, longitudinal observational studies, and clinical trials. The results of animal testing suggest that pioglitazone can be corrective as well as protective, and that its efficacy is enhanced in a time- and dose-dependent manner, but the dose-effect relations are not monotonic or sigmoid. Longitudinal cohort studies suggests that it delays the onset of dementia in individuals with pre-existing type 2 diabetes mellitus, which small scale, unblinded pilot studies seem to confirm. However, the results of placebo-controlled, blinded clinical trials have not borne this out, and we discuss possible explanations for these discrepancies.
Collapse
Affiliation(s)
- Ann M. Saunders
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States
| | - Daniel K. Burns
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States
| | | |
Collapse
|
36
|
Yeh JH, Wang KC, Kaizaki A, Lee JW, Wei HC, Tucci MA, Ojeda NB, Fan LW, Tien LT. Pioglitazone Ameliorates Lipopolysaccharide-Induced Behavioral Impairment, Brain Inflammation, White Matter Injury and Mitochondrial Dysfunction in Neonatal Rats. Int J Mol Sci 2021; 22:6306. [PMID: 34208374 PMCID: PMC8231261 DOI: 10.3390/ijms22126306] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/29/2021] [Accepted: 06/05/2021] [Indexed: 01/04/2023] Open
Abstract
Previous studies have demonstrated that pioglitazone, a peroxisome proliferator-activated receptor gamma (PPARγ) agonist, inhibits ischemia-induced brain injury. The present study was conducted to examine whether pioglitazone can reduce impairment of behavioral deficits mediated by inflammatory-induced brain white matter injury in neonatal rats. Intraperitoneal (i.p.) injection of lipopolysaccharide (LPS, 2 mg/kg) was administered to Sprague-Dawley rat pups on postnatal day 5 (P5), and i.p. administration of pioglitazone (20 mg/kg) or vehicle was performed 5 min after LPS injection. Sensorimotor behavioral tests were performed 24 h after LPS exposure, and changes in biochemistry of the brain was examined after these tests. The results show that systemic LPS exposure resulted in impaired sensorimotor behavioral performance, reduction of oligodendrocytes and mitochondrial activity, and increases in lipid peroxidation and brain inflammation, as indicated by the increment of interleukin-1β (IL-1β) levels and number of activated microglia in the neonatal rat brain. Pioglitazone treatment significantly improved LPS-induced neurobehavioral and physiological disturbances including the loss of body weight, hypothermia, righting reflex, wire-hanging maneuver, negative geotaxis, and hind-limb suspension in neonatal rats. The neuroprotective effect of pioglitazone against the loss of oligodendrocytes and mitochondrial activity was associated with attenuation of LPS-induced increment of thiobarbituric acid reactive substances (TBARS) content, IL-1β levels and number of activated microglia in neonatal rats. Our results show that pioglitazone prevents neurobehavioral disturbances induced by systemic LPS exposure in neonatal rats, and its neuroprotective effects are associated with its impact on microglial activation, IL-1β induction, lipid peroxidation, oligodendrocyte production and mitochondrial activity.
Collapse
Affiliation(s)
- Jiann-Horng Yeh
- Department of Neurobiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan;
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City 24205, Taiwan; (K.-C.W.); (H.-C.W.)
| | - Kuo-Ching Wang
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City 24205, Taiwan; (K.-C.W.); (H.-C.W.)
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan
| | - Asuka Kaizaki
- Department of Pharmacology, Toxicology and Therapeutics, Division of Toxicology, School of Pharmacy, Showa University, Shingawa-ku, Tokyo 142-8555, Japan;
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.L.); (N.B.O.); (L.-W.F.)
| | - Jonathan W. Lee
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.L.); (N.B.O.); (L.-W.F.)
| | - Han-Chi Wei
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City 24205, Taiwan; (K.-C.W.); (H.-C.W.)
| | - Michelle A. Tucci
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Norma B. Ojeda
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.L.); (N.B.O.); (L.-W.F.)
| | - Lir-Wan Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.W.L.); (N.B.O.); (L.-W.F.)
| | - Lu-Tai Tien
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City 24205, Taiwan; (K.-C.W.); (H.-C.W.)
| |
Collapse
|
37
|
Batabyal R, Freishtat N, Hill E, Rehman M, Freishtat R, Koutroulis I. Metabolic dysfunction and immunometabolism in COVID-19 pathophysiology and therapeutics. Int J Obes (Lond) 2021; 45:1163-1169. [PMID: 33727631 PMCID: PMC7961323 DOI: 10.1038/s41366-021-00804-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 02/22/2021] [Accepted: 03/04/2021] [Indexed: 12/23/2022]
Abstract
The COVID-19 pandemic has emerged as a public health crisis and has placed a significant burden on healthcare systems. Patients with underlying metabolic dysfunction, such as type 2 diabetes mellitus and obesity, are at a higher risk for COVID-19 complications, including multi-organ dysfunction, secondary to a deranged immune response, and cellular energy deprivation. These patients are at a baseline state of chronic inflammation associated with increased susceptibility to the severe immune manifestations of COVID-19, which are triggered by the cellular hypoxic environment and cytokine storm. The altered metabolic profile and energy generation of immune cells affect their activation, exacerbating the imbalanced immune response. Key immunometabolic interactions may inform the development of an efficacious treatment for COVID-19. Novel therapeutic approaches with repurposed drugs, such as PPAR agonists, or newly developed molecules such as the antagomirs, which block microRNA function, have shown promising results. Those treatments, alone or in combination, target both immune and metabolic pathways and are ideal for septic COVID-19 patients with an underlying metabolic condition.
Collapse
Affiliation(s)
- Rachael Batabyal
- Division of Emergency Medicine, Children's National Hospital, Washington, DC, USA
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Nathaniel Freishtat
- Division of Emergency Medicine, Children's National Hospital, Washington, DC, USA
| | - Elaise Hill
- Division of Emergency Medicine, Children's National Hospital, Washington, DC, USA
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Muhammad Rehman
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Robert Freishtat
- Division of Emergency Medicine, Children's National Hospital, Washington, DC, USA
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Ioannis Koutroulis
- Division of Emergency Medicine, Children's National Hospital, Washington, DC, USA.
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
38
|
Al-Muzafar HM, Alshehri FS, Amin KA. The role of pioglitazone in antioxidant, anti-inflammatory, and insulin sensitivity in a high fat-carbohydrate diet-induced rat model of insulin resistance. ACTA ACUST UNITED AC 2021; 54:e10782. [PMID: 34037093 PMCID: PMC8148887 DOI: 10.1590/1414-431x2020e10782] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/15/2021] [Indexed: 01/13/2023]
Abstract
We explored the cascade effects of a high fat-carbohydrate diet (HFCD) and pioglitazone (an anti-diabetic therapy used to treat type 2 diabetes mellitus (T2DM)) on lipid profiles, oxidative stress/antioxidant, insulin, and inflammatory biomarkers in a rat model of insulin resistance. Sixty albino rats (80-90 g) were randomly divided into three dietary groups; 1) standard diet; 2) HFCD diet for 12 weeks to induce an in vivo model of insulin resistance; and 3) HFCD diet plus pioglitazone. Blood and tissue samples were taken to assess hepatic function, lipid profiles, oxidative biomarkers, malondialdehyde (MDA) levels, antioxidant defense biomarkers, including reduced glutathione (GSH), superoxide dismutase (SOD), and the inflammatory markers interleukin-6 (IL-6) and tumor necrotic factor (TNF-α). HFCD-fed rats had significantly (P≤0.05) increased serum triacylglycerol (TG), total cholesterol (TC), low-density lipoprotein (LDL), alanine transaminase (ALT), and bilirubin levels, but decreased high-density lipoprotein (HDL) levels compared with the normal group. Moreover, serum leptin, resistin, TNF-α, and IL-6 levels were increased significantly in HFCD animals compared with controls. Similarly, HFCD-induced insulin resistance caused antioxidant and cytokine disturbances, which are important therapy targets for pioglitazone. Importantly, administration of this drug ameliorated these changes, normalized leptin and resistin and inflammatory markers by reducing TNF-α levels. Metabolic cascades of elevated lipid profiles, oxidative stress, insulin, and inflammatory biomarkers are implicated in insulin resistance progression. HFCD induced metabolic cascades comprising hypertriglyceridemia, hyperglycemia, insulin resistance, obesity-associated hormones, and inflammatory biomarkers may be alleviated using pioglitazone.
Collapse
Affiliation(s)
- H M Al-Muzafar
- Department of Chemistry, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.,Basic & Applied Scientific Research Center, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - F S Alshehri
- Department of Chemistry, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.,Basic & Applied Scientific Research Center, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - K A Amin
- Department of Chemistry, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.,Basic & Applied Scientific Research Center, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
39
|
Telek V, Erlitz L, Caleb I, Nagy T, Vecsernyés M, Balogh B, Sétáló G, Hardi P, Jancsó G, Takács I. Effect of Pioglitazone on endoplasmic reticulum stress regarding in situ perfusion rat model. Clin Hemorheol Microcirc 2021; 79:311-325. [PMID: 33867357 DOI: 10.3233/ch-211163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) can cause insufficient microcirculation of the transplanted organ and results in a diminished and inferior graft survival rate. OBJECTIVE This study aimed to investigate the effect of different doses of an anti-diabetic drug, Pioglitazone (Pio), on endoplasmic reticulum stress and histopathological changes, using an in situ perfusion rat model. METHODS Sixty male Wistar rats were used and were divided into six groups, consisting of the control group, vehicle-treated group and four Pio-treated groups (10, 20, 30 and 40 mg/kg Pio was administered). The rats were perfused through vena cava and an outflow on the abdominal aorta occurred. Following the experiment, kidneys and livers were collected. The level of the endoplasmic reticulum stress markers (XBP1 and Caspase 12) was analyzed using Western blot and histopathological changes were evaluated. RESULTS Histopathological findings were correlated with the Western blot results and depict a protective effect corresponding to the elevated dosage of Pioglitazone regarding in situ perfusion rat model. CONCLUSIONS In our study, Pioglitazone can reduce the endoplasmic reticulum stress, and the most effective dosage proved to be the 40 mg/kg Pio referencing the kidney and liver samples.
Collapse
Affiliation(s)
- Vivien Telek
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| | - Luca Erlitz
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| | - Ibitamuno Caleb
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| | - Tibor Nagy
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| | - Mónika Vecsernyés
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Pécs, Hungary
| | - Bálint Balogh
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Pécs, Hungary
| | - György Sétáló
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Pécs, Hungary.,Signal Transduction Research Group, János Szentágothai Research Centre, Pécs, Hungary
| | - Péter Hardi
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| | - Gábor Jancsó
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| | - Ildikó Takács
- Department of Surgical Research and Techniques, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
40
|
de Carvalho MV, Gonçalves-de-Albuquerque CF, Silva AR. PPAR Gamma: From Definition to Molecular Targets and Therapy of Lung Diseases. Int J Mol Sci 2021; 22:E805. [PMID: 33467433 PMCID: PMC7830538 DOI: 10.3390/ijms22020805] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 12/15/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor superfamily that regulate the expression of genes related to lipid and glucose metabolism and inflammation. There are three members: PPARα, PPARβ or PPARγ. PPARγ have several ligands. The natural agonists are omega 9, curcumin, eicosanoids and others. Among the synthetic ligands, we highlight the thiazolidinediones, clinically used as an antidiabetic. Many of these studies involve natural or synthetic products in different pathologies. The mechanisms that regulate PPARγ involve post-translational modifications, such as phosphorylation, sumoylation and ubiquitination, among others. It is known that anti-inflammatory mechanisms involve the inhibition of other transcription factors, such as nuclear factor kB(NFκB), signal transducer and activator of transcription (STAT) or activator protein 1 (AP-1), or intracellular signaling proteins such as mitogen-activated protein (MAP) kinases. PPARγ transrepresses other transcription factors and consequently inhibits gene expression of inflammatory mediators, known as biomarkers for morbidity and mortality, leading to control of the exacerbated inflammation that occurs, for instance, in lung injury/acute respiratory distress. Many studies have shown the therapeutic potentials of PPARγ on pulmonary diseases. Herein, we describe activities of the PPARγ as a modulator of inflammation, focusing on lung injury and including definition and mechanisms of regulation, biological effects and molecular targets, and its role in lung diseases caused by inflammatory stimuli, bacteria and virus, and molecular-based therapy.
Collapse
Affiliation(s)
- Márcia V. de Carvalho
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil;
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil
| | - Cassiano F. Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil;
- Laboratório de Imunofarmacologia, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro 20211-010, Brazil
- Programa de Pós-Graduação em Biologia Molecular e Celular, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro 20211-010, Brazil
| | - Adriana R. Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil;
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
41
|
Nikitovic D. The role of extracellular matrix in allergic contact dermatitis pathogenesis. TOXICOLOGICAL RISK ASSESSMENT AND MULTI-SYSTEM HEALTH IMPACTS FROM EXPOSURE 2021:205-214. [DOI: 10.1016/b978-0-323-85215-9.00012-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
42
|
Cadegiani FA. Repurposing existing drugs for COVID-19: an endocrinology perspective. BMC Endocr Disord 2020; 20:149. [PMID: 32993622 PMCID: PMC7523486 DOI: 10.1186/s12902-020-00626-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Coronavirus Disease 2019 (COVID-19) is a multi-systemic infection caused by the novel Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), that has become a pandemic. Although its prevailing symptoms include anosmia, ageusia, dry couch, fever, shortness of brief, arthralgia, myalgia, and fatigue, regional and methodological assessments vary, leading to heterogeneous clinical descriptions of COVID-19. Aging, uncontrolled diabetes, hypertension, obesity, and exposure to androgens have been correlated with worse prognosis in COVID-19. Abnormalities in the renin-angiotensin-aldosterone system (RAAS), angiotensin-converting enzyme-2 (ACE2) and the androgen-driven transmembrane serine protease 2 (TMPRSS2) have been elicited as key modulators of SARS-CoV-2. MAIN TEXT While safe and effective therapies for COVID-19 lack, the current moment of pandemic urges for therapeutic options. Existing drugs should be preferred over novel ones for clinical testing due to four inherent characteristics: 1. Well-established long-term safety profile, known risks and contraindications; 2. More accurate predictions of clinical effects; 3. Familiarity of clinical management; and 4. Affordable costs for public health systems. In the context of the key modulators of SARS-CoV-2 infectivity, endocrine targets have become central as candidates for COVID-19. The only endocrine or endocrine-related drug class with already existing emerging evidence for COVID-19 is the glucocorticoids, particularly for the use of dexamethasone for severely affected patients. Other drugs that are more likely to present clinical effects despite the lack of specific evidence for COVID-19 include anti-androgens (spironolactone, eplerenone, finasteride and dutasteride), statins, N-acetyl cysteine (NAC), ACE inhibitors (ACEi), angiotensin receptor blockers (ARB), and direct TMPRSS-2 inhibitors (nafamostat and camostat). Several other candidates show less consistent plausibility. In common, except for dexamethasone, all candidates have no evidence for COVID-19, and clinical trials are needed. CONCLUSION While dexamethasone may reduce mortality in severely ill patients with COVID-19, in the absence of evidence of any specific drug for mild-to-moderate COVID-19, researchers should consider testing existing drugs due to their favorable safety, familiarity, and cost profile. However, except for dexamethasone in severe COVID-19, drug treatments for COVID-19 patients must be restricted to clinical research studies until efficacy has been extensively proven, with favorable outcomes in terms of reduction in hospitalization, mechanical ventilation, and death.
Collapse
Affiliation(s)
- Flavio A Cadegiani
- Adrenal and Hypertension Unit, Division of Endocrinology and Metabolism, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/UNIFESP), Rua Pedro de Toledo 781 - 13th floor, São Paulo, SP, 04039-032, Brazil.
| |
Collapse
|
43
|
Schulte R, Wohlleber D, Unrau L, Geers B, Metzger C, Erhardt A, Tiegs G, van Rooijen N, Heukamp LC, Klotz L, Knolle PA, Diehl L. Pioglitazone-Mediated Peroxisome Proliferator-Activated Receptor γ Activation Aggravates Murine Immune-Mediated Hepatitis. Int J Mol Sci 2020; 21:ijms21072523. [PMID: 32260486 PMCID: PMC7177299 DOI: 10.3390/ijms21072523] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/20/2022] Open
Abstract
The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) regulates target gene expression upon ligand binding. Apart from its effects on metabolism, PPARγ activity can inhibit the production of pro-inflammatory cytokines by several immune cells, including dendritic cells and macrophages. In chronic inflammatory disease models, PPARγ activation delays the onset and ameliorates disease severity. Here, we investigated the effect of PPARγ activation by the agonist Pioglitazone on the function of hepatic immune cells and its effect in a murine model of immune-mediated hepatitis. Cytokine production by both liver sinusoidal endothelial cells (IL-6) and in T cells ex vivo (IFNγ) was decreased in cells from Pioglitazone-treated mice. However, PPARγ activation did not decrease pro-inflammatory tumor necrosis factor alpha TNFα production by Kupffer cells after Toll-like receptor (TLR) stimulation ex vivo. Most interestingly, although PPARγ activation was shown to ameliorate chronic inflammatory diseases, it did not improve hepatic injury in a model of immune-mediated hepatitis. In contrast, Pioglitazone-induced PPARγ activation exacerbated D-galactosamine (GalN)/lipopolysaccharide (LPS) hepatitis associated with an increased production of TNFα by Kupffer cells and increased sensitivity of hepatocytes towards TNFα after in vivo Pioglitazone administration. These results unravel liver-specific effects of Pioglitazone that fail to attenuate liver inflammation but rather exacerbate liver injury in an experimental hepatitis model.
Collapse
Affiliation(s)
- Rike Schulte
- Institute for Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany; (R.S.); (D.W.); (C.M.); (L.K.); (P.A.K.)
| | - Dirk Wohlleber
- Institute for Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany; (R.S.); (D.W.); (C.M.); (L.K.); (P.A.K.)
- Institute of Molecular Immunology and Experimental Oncology, Technical University Munich, 81675, Munich, Germany
| | - Ludmilla Unrau
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.U.); (B.G); (A.E.); (G.T.)
| | - Bernd Geers
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.U.); (B.G); (A.E.); (G.T.)
| | - Christina Metzger
- Institute for Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany; (R.S.); (D.W.); (C.M.); (L.K.); (P.A.K.)
| | - Annette Erhardt
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.U.); (B.G); (A.E.); (G.T.)
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.U.); (B.G); (A.E.); (G.T.)
| | - Nico van Rooijen
- Department of Molecular Cell Biology, VU University Medical Center, 1081 HV Amsterdam, The Netherlands;
| | | | - Luisa Klotz
- Institute for Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany; (R.S.); (D.W.); (C.M.); (L.K.); (P.A.K.)
- Department of Neurology, University Hospital Münster, 48149 Münster, Germany
| | - Percy A. Knolle
- Institute for Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany; (R.S.); (D.W.); (C.M.); (L.K.); (P.A.K.)
- Institute of Molecular Immunology and Experimental Oncology, Technical University Munich, 81675, Munich, Germany
| | - Linda Diehl
- Institute for Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany; (R.S.); (D.W.); (C.M.); (L.K.); (P.A.K.)
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.U.); (B.G); (A.E.); (G.T.)
- Correspondence:
| |
Collapse
|
44
|
Thioredoxin-2 impacts the inflammatory response via suppression of NF-κB and MAPK signaling in sepsis shock. Biochem Biophys Res Commun 2020; 524:876-882. [PMID: 32057359 DOI: 10.1016/j.bbrc.2020.01.169] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 01/31/2020] [Indexed: 12/24/2022]
Abstract
Sepsis is a progressive disease characterized by excessive inflammatory responses, severe tissue injury and organ dysfunction, ultimately leading to mortality. In this study, we demonstrated that thioredoxin-2 (TRX-2) expression is reduced in macrophages stimulated with lipopolysaccharide (LPS). Overexpression of TRX-2 significantly attenuated interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) production induced by LPS. TRX-2 inhibited LPS-induced inflammatory responses through suppressing activation of the NF-κB and MAPK signaling pathways. Furthermore, TRX-2 induced a significant decrease in mortality in mouse sepsis models in association with reduced inflammatory cytokine production and attenuation of organ injury. Our data collectively support a role of TRX-2 as a critical regulator of sepsis that influences survival by protecting the host from excessive inflammatory damage.
Collapse
|
45
|
Fu J, Li G, Wu X, Zang B. Sodium Butyrate Ameliorates Intestinal Injury and Improves Survival in a Rat Model of Cecal Ligation and Puncture-Induced Sepsis. Inflammation 2020; 42:1276-1286. [PMID: 30903547 DOI: 10.1007/s10753-019-00987-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sepsis is a life-threatening condition with a high rate of mortality. Unfortunately, very few therapies can improve outcomes in patients with sepsis. Butyrate, which is the most potent histone deacetylase (HDAC) inhibitor among short-chain fatty acids, exerts anti-inflammatory effects in a variety of inflammatory diseases. Butyrate might thus be valuable in the treatment of sepsis, in which inhibition of overwhelming cytokine release is vitally important. Sepsis was induced in 7- to 8-week-old Sprague-Dawley rats by cecal ligation and puncture (CLP) with a 21-g double-puncture technique. Rats received an intravenous injection of normal saline (vehicle) or sodium butyrate (200 mg/kg) after CLP and were sacrificed 12 h later. Hematoxylin and eosin staining was performed to observe the intestinal mucosal morphology. RT-PCR and ELISA were used to determine the intestinal inflammatory response in vivo. Intestinal permeability was evaluated by measuring fluorescein isothiocyanate dextran (FD-4) absorption in vivo, and tight junction protein expression was examined by western blot. NF-κB p65 activities were assessed by western blot and immunohistochemistry. Sodium butyrate treatment improved the survival rate of CLP rats and alleviated sepsis-induced intestinal mucosal injury. Proinflammatory cytokine expression was lower in butyrate-treated rats than in the vehicle group. FD-4 leakage from the intestinal tract was reduced, and the expression levels of the tight junction proteins claudin-1 and ZO-1 were also restored in rats that received sodium butyrate treatment. These effects were associated with less NF-κB p65 nuclear translocation, whereas the expression of Iκ-Bα was not affected or even increased. Sodium butyrate mitigates the inflammatory response and maintains intestinal barrier function in polymicrobial sepsis partly through inhibition of NF-κB activation and may serve as a novel therapy for sepsis.
Collapse
Affiliation(s)
- Jiahong Fu
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Guofu Li
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Xingmao Wu
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Bin Zang
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
46
|
Liu J, Huang X, Hu S, He H, Meng Z. Dexmedetomidine attenuates lipopolysaccharide induced acute lung injury in rats by inhibition of caveolin-1 downstream signaling. Biomed Pharmacother 2019; 118:109314. [DOI: 10.1016/j.biopha.2019.109314] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 07/27/2019] [Accepted: 07/31/2019] [Indexed: 10/26/2022] Open
|
47
|
Fan H, Zhao Y, Zhu JH. S-nitrosoglutathione protects lipopolysaccharide-induced acute kidney injury by inhibiting toll-like receptor 4-nuclear factor-κB signal pathway. ACTA ACUST UNITED AC 2019; 71:1255-1261. [PMID: 31115903 DOI: 10.1111/jphp.13103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/22/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVES To investigate the therapeutic effects and mechanisms of S-nitrosoglutathione (SNG) on acute kidney injury (AKI) induced by lipopolysaccharide (LPS). METHODS We established an AKI model by intraperitoneal administration of LPS in mice and LPS-induced human kidney (HK-2) cells in vitro. We obtained the kidney tissues from mice for histopathological examination, examined inflammatory cytokines by enzyme-linked immunosorbent assay and measured the expression levels of toll-like receptor 4-nuclear factor-κB (TLR4-NF-κB) signal pathway-related proteins by Western blotting. KEY FINDINGS Pretreatment of SNG effectively improved the kidney function, reduced the pathological damage score of kidney in mice and decreased the expression levels of IL-1β, IL-6 and TNF-α in a dose-dependent manner in vivo and in vitro. Furthermore, pretreatment of SNG also repressed TLR4, phosphorylated NF-κB IκBα, IKKβ and p65 expression levels in HK-2 cells induced by LPS. CONCLUSIONS S-nitrosoglutathione attenuates the severity of LPS-induced AKI by inhibiting the TLR4-NF-κB signalling pathway and may act as a protective agent for septic AKI.
Collapse
Affiliation(s)
- Heng Fan
- Department of Intensive Care Unit, Ningbo First Hospital, Ningbo, China
| | - Yu Zhao
- Department of Nephrology, Ningbo Urology and Nephrology Hospital, Ningbo, China
| | - Jian-Hua Zhu
- Department of Intensive Care Unit, Ningbo First Hospital, Ningbo, China
| |
Collapse
|
48
|
Nakashima M, Kinoshita M, Nakashima H, Kotani A, Ishikiriyama T, Kato S, Hiroi S, Seki S. Pioglitazone improves phagocytic activity of liver recruited macrophages in elderly mice possibly by promoting glucose catabolism. Innate Immun 2019; 25:356-368. [PMID: 31096821 PMCID: PMC7103614 DOI: 10.1177/1753425919849620] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent studies have revealed that the immunological function of leukocytes is dependent on their cellular metabolism, and some researchers have advocated the beneficial effects of pioglitazone against sepsis in young mice, although bacterial infections are more prevalent in elderly hosts. Here, we investigated pioglitazone’s preventative effect against sepsis induced by intravenous injection of a lethal dose of Escherichia coli in elderly mice (50–60 wk old) and examined its immunological and metabolic effects on liver leukocytes. Pioglitazone improved bacterial elimination in the peripheral blood, lowered serum pro-inflammatory cytokines (TNF-α, IL-12, IFN-γ), and prevented septic death. It also enhanced bacterial elimination in the liver, by increasing the phagocytic and bactericidal activities of liver F4/80+CD11b+ recruited macrophages (Mφ), their CD206 expression and reactive oxygen species production. Quantitative PCR revealed that pioglitazone treatment enhanced gene expression of rate-limiting enzymes for glycolysis in hepatic CD11b+ cells (including neutrophils and recruited Mφ), and their improved phagocytic and bactericidal activities were abolished by glycolysis inhibiting reagents. These findings present the possibility that pioglitazone strengthens the phagocytic and bactericidal activities of liver recruited Mφ and that these immunological activities are closely associated with their glucose catabolism.
Collapse
Affiliation(s)
- Masahiro Nakashima
- 1 Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Japan
| | - Manabu Kinoshita
- 1 Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Japan
| | - Hiroyuki Nakashima
- 1 Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Japan
| | - Aya Kotani
- 1 Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Japan
| | - Takuya Ishikiriyama
- 1 Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Japan
| | - Shoichiro Kato
- 2 Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Sadayuki Hiroi
- 3 Department of Pathology, Nitobebunka College, Nakano, Tokyo, Japan
| | - Shuhji Seki
- 1 Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Japan
| |
Collapse
|
49
|
Silva AR, Gonçalves-de-Albuquerque CF, Pérez AR, Carvalho VDF. Immune-endocrine interactions related to a high risk of infections in chronic metabolic diseases: The role of PPAR gamma. Eur J Pharmacol 2019; 854:272-281. [PMID: 30974105 DOI: 10.1016/j.ejphar.2019.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 03/11/2019] [Accepted: 04/02/2019] [Indexed: 02/06/2023]
Abstract
Diverse disturbances in immune-endocrine circuitries are involved in the development and aggravation of several chronic metabolic diseases (CMDs), including obesity, diabetes, and metabolic syndrome. The chronic inflammatory syndrome observed in CMDs culminates in dysregulated immune responses with low microbial killing efficiency, by means low host innate immune response, and loss of ability to eliminate the pathogens, which results in a high prevalence of infectious diseases, including pneumonia, tuberculosis, and sepsis. Herein, we review evidence pointing out PPARγ as a putative player in immune-endocrine disturbances related to increased risk of infections in CMDs. Cumulated evidence indicates that PPARγ activation modulates host cells to control inflammation during CMDs because of PPARγ agonists have anti-inflammatory and pro-resolutive properties, increasing host ability to eliminate pathogen, modulating hormone production, and restoring glucose and lipid homeostasis. As such, we propose PPARγ as a putative therapeutic adjuvant for patients with CMDs to favor a better infection control.
Collapse
Affiliation(s)
- Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil, 4365, Rio de Janeiro, RJ, Brazil.
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil, 4365, Rio de Janeiro, RJ, Brazil; Laboratório de Imunofarmacologia, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Unirio, Brazil.
| | - Ana Rosa Pérez
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET UNR), 2000, Rosario, Argentina.
| | - Vinicius de Frias Carvalho
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Avenida Brasil, 4365, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
50
|
Bombicz M, Priksz D, Gesztelyi R, Kiss R, Hollos N, Varga B, Nemeth J, Toth A, Papp Z, Szilvassy Z, Juhasz B. The Drug Candidate BGP-15 Delays the Onset of Diastolic Dysfunction in the Goto-Kakizaki Rat Model of Diabetic Cardiomyopathy. Molecules 2019; 24:molecules24030586. [PMID: 30736394 PMCID: PMC6384948 DOI: 10.3390/molecules24030586] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/24/2019] [Accepted: 02/05/2019] [Indexed: 01/01/2023] Open
Abstract
Background and Aims: Diabetic cardiomyopathy (DCM) is an emerging problem worldwide due to an increase in the incidence of type 2 diabetes. Animal studies have indicated that metformin and pioglitazone can prevent DCM partly by normalizing insulin resistance, and partly by other, pleiotropic mechanisms. One clinical study has evidenced the insulin-senzitizing effect of the drug candidate BGP-15, along with additional animal studies that have confirmed its beneficial effects in models of diabetes, muscular dystrophy and heart failure, with the drug affecting chaperones, contractile proteins and mitochondria. Our aim was to investigate whether the inzulin-senzitizer BGP-15 exert any additive cardiovascular effects compared to metformin or pioglitazone, using Goto-Kakizaki (GotoK) rats. Methods: Rats were divided into five groups: (I) healthy control (Wistar), (II) diseased (GotoK), and GotoK rats treated with: (III) BGP-15, (IV) metformin, and (V) pioglitazone, respectively, for 12 weeks. Metabolic parameters and insulin levels were determined at the endpoint. Doppler echocardiography was carried out to estimate diabetes-associated cardiac dysfunction. Thoracotomy was performed after the vascular status of rats was evaluated using an isolated aortic ring method. Furthermore, western blot assays were carried out to determine expression or phosphorylation levels of selected proteins that take part in myocyte relaxation. Results: BGP-15 restored diastolic parameters (e′/a′, E/e′, LAP, E and A wave) and improved Tei-index compared to untreated GotoK rats. Vascular status was unaffected by BGP-15. Expression of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a) and phosphodiesterase 9A (PDE9A) were unchanged by the treatments, but the phosphorylation level of vasodilator-stimulated phosphoprotein (VASP) and phospholamban (PLB) increased in BGP-15-treated rats, in comparison to GotoK. Conclusions: Even though the BGP-15-treatment did not interfere significantly with glucose homeostasis and vascular status, it considerably enhanced diastolic function, by affecting the SERCA/phospholamban pathway in GotoK rats. Although it requires further investigation, BGP-15 may offer a new therapeutic approach in DCM.
Collapse
Affiliation(s)
- Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Daniel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Rudolf Gesztelyi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Rita Kiss
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Nora Hollos
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Balazs Varga
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Jozsef Nemeth
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Attila Toth
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Zoltan Papp
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Zoltan Szilvassy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Bela Juhasz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| |
Collapse
|