1
|
Samantasinghar A, Sunildutt N, Ahmed F, Memon FH, Kang C, Choi KH. Revolutionizing Biomedical Research: Unveiling the Power of Microphysiological Systems with Advanced Assays, Integrated Sensor Technologies, and Real-Time Monitoring. ACS OMEGA 2025; 10:9869-9889. [PMID: 40124012 PMCID: PMC11923667 DOI: 10.1021/acsomega.4c11227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 03/25/2025]
Abstract
The limitation of animal models to imitate a therapeutic response in humans is a key problem that challenges their use in fundamental research. Organ-on-a-chip (OOC) devices, also called microphysiological systems (MPS), are devices containing a lining of living cells grown under dynamic flow to recapitulate the important features of human physiology and pathophysiology with high precision. Recent advances in microfabrication and tissue engineering techniques have led to the wide adoption of OOC in next-generation experimental platforms. This review presents a comprehensive analysis of the OOC systems, categorizing them by flow types (single-pass and multipass), operational mechanisms (pumpless and pump-driven), and configurations (single-organ and multiorgan systems), along with their respective advantages and limitations. Furthermore, it explores the integration of qualitative and quantitative assay techniques, providing a comparative evaluation of systems with and without sensor integration. This review aims to fill essential knowledge gaps, driving the progress of the development of OOC systems and paving the way for breakthroughs in biomedical research, pharmaceutical innovation, and tissue engineering.
Collapse
Affiliation(s)
- Anupama Samantasinghar
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
| | - Naina Sunildutt
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
| | - Faheem Ahmed
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
| | - Fida Hussain Memon
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
- Department
of Electrical Engineering, Sukkur IBA University, Sindh 65200, Pakistan
| | - Chulung Kang
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
| | - Kyung Hyun Choi
- Department
of Mechatronics Engineering, Jeju National
University, Jeju 63243, Republic
of Korea
| |
Collapse
|
2
|
Li J, Guo H, Tan L, Chen M, Wang X, Liu Y, Chen S, Wang Y, Yu H, Wang P. Terahertz Irradiation Promotes Angiogenesis in vitro by Enhancing Permeability of the Voltage-Gated Calcium Channel. PLoS One 2025; 20:e0317426. [PMID: 39982926 PMCID: PMC11844849 DOI: 10.1371/journal.pone.0317426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/29/2024] [Indexed: 02/23/2025] Open
Abstract
Terahertz (THz) waves, positioned between microwave and infrared in the electromagnetic spectrum, have promising applications in medical imaging and biomedicine. In this study, terahertz irradiation at 2.52 THz (100 mW/cm2) did not alter the proliferation of human umbilical vein endothelial cells (HUVECs), but significantly enhanced their angiogenic capacity. This enhancement was accompanied by increased levels of angiogenesis-related proteins such as VEGF in the culture supernatant. ATAC sequencing and RNA sequencing revealed a significant increase in the expression of cytoskeleton-associated genes, including PDXP and SH3BP1, post-irradiation. Additionally, intracellular calcium concentration, closely linked to angiogenesis, markedly increased following terahertz exposure. However, diltiazem significantly mitigated the enhanced angiogenic capacity induced by terahertz irradiation. In conclusion, terahertz irradiation promotes angiogenesis in HUVECs, partly by activating the VEGF signaling pathway through increased calcium fluxes.
Collapse
Affiliation(s)
- Jichang Li
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, China
| | - Hongjie Guo
- Division of Vascular Interventional, Peking University First Hospital, Beijing, China
| | - Linxia Tan
- National Engineering Research Center for Dangerous Articles and Explosives Detection Technologies, Department of Engineering Physics, Tsinghua University, Beijing, China
- Weimai Qingtong Medical Technology Co., Wuxi, China
| | - Meng Chen
- National Engineering Research Center for Dangerous Articles and Explosives Detection Technologies, Department of Engineering Physics, Tsinghua University, Beijing, China
| | - Xin Wang
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, China
| | - Yucun Liu
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, China
| | - Shanwen Chen
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, China
| | - Yingxin Wang
- National Engineering Research Center for Dangerous Articles and Explosives Detection Technologies, Department of Engineering Physics, Tsinghua University, Beijing, China
| | - Hao Yu
- Weimai Qingtong Medical Technology Co., Wuxi, China
| | - Pengyuan Wang
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
3
|
Liu H, Magaye R, Kaye DM, Wang BH. Heart failure with preserved ejection fraction: The role of inflammation. Eur J Pharmacol 2024; 980:176858. [PMID: 39074526 DOI: 10.1016/j.ejphar.2024.176858] [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: 02/22/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Heart failure (HF) is a debilitating clinical syndrome affecting 64.3 million patients worldwide. More than 50% of HF cases are attributed to HF with preserved ejection fraction (HFpEF), an entity growing in prevalence and mortality. Although recent breakthroughs reveal the prognostic benefits of sodium-glucose co-transporter 2 inhibitors (SGLT2i) in HFpEF, there is still a lack of effective pharmacological therapy available. This highlights a major gap in medical knowledge that must be addressed. Current evidence attributes HFpEF pathogenesis to an interplay between cardiometabolic comorbidities, inflammation, and renin-angiotensin-aldosterone-system (RAAS) activation, leading to cardiac remodelling and diastolic dysfunction. However, conventional RAAS blockade has demonstrated limited benefits in HFpEF, which emphasises that alternative therapeutic targets should be explored. Presently, there is limited literature examining the use of anti-inflammatory HFpEF therapies despite growing evidence supporting its importance in disease progression. Hence, this review aims to explore current perspectives on HFpEF pathogenesis, including the importance of inflammation-driven cardiac remodelling and the therapeutic potential of anti-inflammatory therapies.
Collapse
Affiliation(s)
- Hongyi Liu
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia; Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - Ruth Magaye
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - David M Kaye
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - Bing H Wang
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia; Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.
| |
Collapse
|
4
|
Cazzola R, Della Porta M, Piuri G, Maier JA. Magnesium: A Defense Line to Mitigate Inflammation and Oxidative Stress in Adipose Tissue. Antioxidants (Basel) 2024; 13:893. [PMID: 39199139 PMCID: PMC11351329 DOI: 10.3390/antiox13080893] [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: 06/28/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 09/01/2024] Open
Abstract
Magnesium (Mg) is involved in essential cellular and physiological processes. Globally, inadequate consumption of Mg is widespread among populations, especially those who consume processed foods, and its homeostasis is impaired in obese individuals and type 2 diabetes patients. Since Mg deficiency triggers oxidative stress and chronic inflammation, common features of several frequent chronic non-communicable diseases, interest in this mineral is growing in clinical medicine as well as in biomedicine. To date, very little is known about the role of Mg deficiency in adipose tissue. In obesity, the increase in fat tissue leads to changes in the release of cytokines, causing low-grade inflammation and macrophage infiltration. Hypomagnesemia in obesity can potentiate the excessive production of reactive oxygen species, mitochondrial dysfunction, and decreased ATP production. Importantly, Mg plays a role in regulating intracellular calcium concentration and is involved in carbohydrate metabolism and insulin receptor activity. This narrative review aims to consolidate existing knowledge, identify research gaps, and raise awareness of the critical role of Mg in supporting adipose tissue metabolism and preventing oxidative stress.
Collapse
Affiliation(s)
| | | | | | - Jeanette A. Maier
- Department of Biomedical and Clinical Sciences, University of Milano, 20174 Milan, Italy; (R.C.); (M.D.P.); (G.P.)
| |
Collapse
|
5
|
Li J, Zhou K, Chen X, Lu X, Kong D. Correlation between serum leptin level and sleep monitoring indexes in patients with obstructive sleep apnea hypopnea syndrome and its predictive value: a cross-sectional analysis. Front Med (Lausanne) 2024; 11:1346195. [PMID: 38711782 PMCID: PMC11070583 DOI: 10.3389/fmed.2024.1346195] [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: 11/29/2023] [Accepted: 03/18/2024] [Indexed: 05/08/2024] Open
Abstract
Objective To investigate the association between serum leptin (LP) level and polysomnography (PSG) parameters in patients with obstructive sleep apnea hypopnea syndrome (OSAHS). Methods A cross-sectional study was conducted. The data of subjects who underwent PSG at hospital between January 2021 and December 2022 were collected retrospectively, 220 participants were included. The subjects were categorized into simple snoring group (n = 45), mild OSAHS group (n = 63), moderate OSAHS group (n = 52), and severe OSAHS group (n = 60). The general characteristics, PSG indices, and serological indices were collected retrospectively. Pearson correlation analysis was used to observe the correlation between serum LP level and PSG parameters. The value of serum LP level in predicting OSAHS was analyzed by receiver operating characteristic curve. Results The serum LP level was positively correlated with micro-arousal count, micro-arousal index (MAI), high apnea hypopnea index, times of blood oxygen decreased by≥3% and time in saturation lower 90%, and negatively correlated with lowest nocturnal oxygen saturation and mean oxygen saturation (p < 0.05). The area under the curve (AUC) of serum LP level in predicting the occurrence of OSAHS was 0.8276 (95% CI: 0.7713-0.8839), and when the Youden index was 0.587, the sensitivity was 72.00%, and the specificity was 86.67% (p < 0.0001). In the population with high MAI, the AUC of serum LP level in predicting the occurrence of OSAHS was 0.8825 (95% CI: 0.7833-0.9817), and when the Youden index was 0.690, the sensitivity was 79.00% and the specificity was 90.00% (p < 0.0001). Conclusion Serum LP level is associated with the severity of OSAHS. Serum LP level demonstrates a strong predictive value for the occurrence of OSAHS, particularly in population with high MAI.
Collapse
Affiliation(s)
- Ji Li
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Kejing Zhou
- Department of Ophthalmology, Ningbo Yinzhou No.2 Hospital, Ningbo, Zhejiang, China
| | - Xing Chen
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xu Lu
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Deqiu Kong
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
6
|
Al-Mutairy EA, Al Qattan S, Khalid M, Al-Enazi AA, Al-Saif MM, Imtiaz F, Ramzan K, Raveendran V, Alaiya A, Meyer BF, Atamas SP, Collison KS, Khabar KS, Hasday JD, Al-Mohanna F. Wild-type S100A3 and S100A13 restore calcium homeostasis and mitigate mitochondrial dysregulation in pulmonary fibrosis patient-derived cells. Front Cell Dev Biol 2023; 11:1282868. [PMID: 38099297 PMCID: PMC10720433 DOI: 10.3389/fcell.2023.1282868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
Patients with digenic S100A3 and S100A13 mutations exhibited an atypical and progressive interstitial pulmonary fibrosis, with impaired intracellular calcium homeostasis and mitochondrial dysfunction. Here we provide direct evidence of a causative effect of the mutation on receptor mediated calcium signaling and calcium store responses in control cells transfected with mutant S100A3 and mutant S100A13. We demonstrate that the mutations lead to increased mitochondrial mass and hyperpolarization, both of which were reversed by transfecting patient-derived cells with the wild type S100A3 and S100A13, or extracellular treatment with the recombinant proteins. In addition, we demonstrate increased secretion of inflammatory mediators in patient-derived cells and in control cells transfected with the mutant-encoding constructs. These findings indicate that treatment of patients' cells with recombinant S100A3 and S100A13 proteins is sufficient to normalize most of cellular responses, and may therefore suggest the use of these recombinant proteins in the treatment of this devastating disease.
Collapse
Affiliation(s)
- Eid A. Al-Mutairy
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Somaya Al Qattan
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mohammed Khalid
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Azizah A. Al-Enazi
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Maher M. Al-Saif
- BioMolecular Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Faiqa Imtiaz
- Clinical Genomics, Center of Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Khushnooda Ramzan
- Clinical Genomics, Center of Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Vineesh Raveendran
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ayodele Alaiya
- Stem Cell Therapy Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Brian F. Meyer
- Clinical Genomics, Center of Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Sergei P. Atamas
- University of Maryland School of Medicine, Baltimore, MD, United States
- Baltimore VA Medical Center, Baltimore, MD, United States
| | - Kate S. Collison
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Khalid S. Khabar
- BioMolecular Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Jeffrey D. Hasday
- University of Maryland School of Medicine, Baltimore, MD, United States
- Baltimore VA Medical Center, Baltimore, MD, United States
| | - Futwan Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Tang Q, Yu R, Wang Y, Xie F, Zhang H, Wu C, Fang M. Varied hypoxia adaptation patterns of embryonic brain at different development stages between Tibetan and Dwarf laying chickens. BMC Genomics 2023; 24:342. [PMID: 37344809 PMCID: PMC10286358 DOI: 10.1186/s12864-023-09457-4] [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: 02/10/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Tibetan chickens (Gallus gallus; TBCs), an indigenous breed distributed in the Qinghai-Tibet Plateau, are well adapted to the hypoxic environment. Currently, the molecular genetic basis of hypoxia adaptation in TBCs remains unclear. This study investigated hypoxia adaptation patterns of embryonic brain at different development stages by integrating analysis of the transcriptome with our previously published metabolome data in TBCs and Dwarf Laying Chickens (DLCs), a lowland chicken breed. RESULTS During hypoxia, the results revealed that 1334, 578, and 417 differentially expressed genes (DEGs) (|log2 fold change|>1, p-value < 0.05) on days 8, 12, and 18 of development, respectively between TBCs and DLCs. Gene Ontology (GO) and pathway analyses revealed that DEGs are mainly related to metabolic pathways, vessel development, and immune response under hypoxia. This is consistent with our metabolome data that TBCs have higher energy metabolism than DLCs during hypoxia. Some vital DEGs between TBCs and DLCs, such as EPAS1, VEGFD, FBP1, FBLN5, LDHA, and IL-6 which are involved in the HIF pathway and hypoxia regulation. CONCLUSION These results suggest varied adaptation patterns between TBCs and DLCs under hypoxia. Our study provides a basis for uncovering the molecular regulation mechanism of hypoxia adaptation in TBCs and a potential application of hypoxia adaptation research for other animals living on the Qinghai-Tibet Plateau, and may even contribute to the study of brain diseases caused by hypoxia.
Collapse
Affiliation(s)
- Qiguo Tang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China.
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, China.
| | - Runjie Yu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Yubei Wang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Fuyin Xie
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Hao Zhang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Changxin Wu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Meiying Fang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, China.
| |
Collapse
|
8
|
Skrypnik D, Skrypnik K, Suliburska J, Bogdański P. Leptin-VEGF crosstalk in excess body mass and related disorders: A systematic review. Obes Rev 2023:e13575. [PMID: 37230803 DOI: 10.1111/obr.13575] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/17/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023]
Abstract
By 2030, it is expected that a billion people will have suffer from obesity. Adipose tissue synthesizes leptin, an adipokine that affects cardiovascular risk. Leptin intensifies the synthesis of vascular endothelial growth factor (VEGF). Our study reviews recent reports on leptin-VEGF crosstalk in obesity and related disorders. PubMed, Web of Science, Scopus, and Google Scholar were searched. One hundred and one articles involving human, animal, and in vitro research were included. In vitro studies show the crucial role of interaction between endothelial cells and adipocytes and hypoxia as a factor that intensifies leptin's effects on VEGF. Leptin-VEGF crosstalk promotes the progression of cancer. The animal research reveal that a high-fat diet enhances leptin and VEGF crosstalk. Genetic and epigenetic mechanisms and procreator-offspring programming may be involved in leptin-VEGF crosstalk. Some female-specific characteristics of leptin-VEGF relation in obesity were observed. The human studies have shown that increased leptin and VEGF synthesis and leptin-VEGF crosstalk are factors linking obesity with elevated cardiovascular risk. The studies of the last 10 years documented a range of significant aspects of leptin-VEGF crosstalk specific for obesity and related disorders, shedding new light on the link between obesity and increased cardiovascular risk.
Collapse
Affiliation(s)
- Damian Skrypnik
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznań University of Medical Sciences, Poznań, Poland
| | - Katarzyna Skrypnik
- Department of Human Nutrition and Dietetics, Poznań University of Life Sciences, Poznań, Poland
| | - Joanna Suliburska
- Department of Human Nutrition and Dietetics, Poznań University of Life Sciences, Poznań, Poland
| | - Paweł Bogdański
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|
9
|
Missawi O, Jeddou IB, Venditti M, Zitouni N, Zaouali MA, Abdennebi HB, Messaoudi I, Reiter RJ, Minucci S, Banni M. Environmental microplastic accumulation exacerbates liver ischemia-reperfusion injury in rat: Protective effects of melatonin. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 860:160155. [PMID: 36436653 DOI: 10.1016/j.scitotenv.2022.160155] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/18/2022] [Accepted: 11/09/2022] [Indexed: 06/16/2023]
Abstract
Ischemia-reperfusion (IR) injury is an inevitable complication of liver transplantation and partial hepatectomy. Although the hazards of environmental microplastics (EMPs) have been well explored, data underlying their impact on IR-induced hepatotoxicity and how to alleviate these damages remain largely undefined. In this study, the involvement of melatonin (MT) in modulating EMPs toxicity in the liver undergoing ischemia-reperfusion injury was investigated. Male Wistar rats were exposed to MPs for 7 days and then subjected to 1 h of partial warm ischemia (70 %) followed by 24 h of reperfusion. We analyzed some parameters as the oxidative stress, the stability of cytoskeleton as well as inflammation, and autophagy. Our data suggested that EMPs elicited liver injury in ischemic animals. Data revealed several histological alterations caused by EMP and IRI, including cellular disorientation, cell necrosis, and microvacuolar steatosis, as well as inflammatory cell infiltration. EMPs increased blood transaminase (AST and ALT) and oxidative stress levels in the ischemic liver. In addition, RT-qPCR, immunofluorescence, and western blot analyses highlighted an increased expression of α-tubulin, IL-18, NFkB, and LC3. However, the ability of MT to reduce MPs and IRI toxicity was consistent with a significant decrease in the evaluated markers. The combined data not only document that melatonin is an effective agent to protect against hepatic IRI but also reduces cellular dysfunction caused by EMPs.
Collapse
Affiliation(s)
- Omayma Missawi
- Laboratory of Agrobiodiversity and Ecotoxicology LR21AGR02, ISA Chott-Mariem, Sousse University, Tunisia; Higher Institute of Biotechnology, Monastir University, Monastir, Tunisia
| | - Ikram Ben Jeddou
- Laboratory of Human Genome and multifactorial Diseases (LR12ES07), Faculty of Pharmacie of Monastisr, Monastir University, Tunisia
| | - Massimo Venditti
- Department of Experimental Medicine, Section Human Physiology and Integrated Biological Functions "F. Bottazzi", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Nesrine Zitouni
- Laboratory of Agrobiodiversity and Ecotoxicology LR21AGR02, ISA Chott-Mariem, Sousse University, Tunisia; Higher Institute of Biotechnology, Monastir University, Monastir, Tunisia
| | - Mohamed Amin Zaouali
- Laboratory of Human Genome and multifactorial Diseases (LR12ES07), Faculty of Pharmacie of Monastisr, Monastir University, Tunisia
| | - Hassen Ben Abdennebi
- Laboratory of Human Genome and multifactorial Diseases (LR12ES07), Faculty of Pharmacie of Monastisr, Monastir University, Tunisia
| | - Imed Messaoudi
- LR11ES41, Higher Institute of Biotechnology, Monastir University, 5000 Monastir, Tunisia
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, Joe R. and Teresa Lozano Long School of Medicine, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Sergio Minucci
- Department of Experimental Medicine, Section Human Physiology and Integrated Biological Functions "F. Bottazzi", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mohamed Banni
- Laboratory of Agrobiodiversity and Ecotoxicology LR21AGR02, ISA Chott-Mariem, Sousse University, Tunisia; Higher Institute of Biotechnology, Monastir University, Monastir, Tunisia.
| |
Collapse
|
10
|
Shan Y, Chen Y, Gu H, Wang Y, Sun Y. Regulatory Basis of Adipokines Leptin and Adiponectin in Epilepsy: from Signaling Pathways to Glucose Metabolism. Neurochem Res 2023; 48:2017-2028. [PMID: 36797447 PMCID: PMC10181973 DOI: 10.1007/s11064-023-03891-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/25/2023] [Accepted: 02/07/2023] [Indexed: 02/18/2023]
Abstract
Epilepsy is a common and severe neurological disorder in which impaired glucose metabolism leads to changes in neuronal excitability that slow or promote the development of epilepsy. Leptin and adiponectin are important mediators regulating glucose metabolism in the peripheral and central nervous systems. Many studies have reported a strong association between epilepsy and these two adipokines involved in multiple signaling cascades and glucose metabolism. Due to the complex regulatory mechanisms between them and various signal activation networks, their role in epilepsy involves many aspects, including the release of inflammatory mediators, oxidative damage, and neuronal apoptosis. This paper aims to summarize the signaling pathways involved in leptin and adiponectin and the regulation of glucose metabolism from the perspective of the pathogenesis of epilepsy. In particular, we discuss the dual effects of leptin in epilepsy and the relationship between antiepileptic drugs and changes in the levels of these two adipokines. Clinical practitioners may need to consider these factors in evaluating clinical drugs. Through this review, we can better understand the specific involvement of leptin and adiponectin in the pathogenesis of epilepsy, provide ideas for further exploration, and bring about practical significance for the treatment of epilepsy, especially for the development of personalized treatment according to individual metabolic characteristics.
Collapse
Affiliation(s)
- Yisi Shan
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China.,Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China
| | - Yeting Chen
- Department of Acupuncture, Zhangjiagang Second People's Hospital, Zhangjiagang, 215600, China
| | - Haiping Gu
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China
| | - Yadong Wang
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China
| | - Yaming Sun
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China.
| |
Collapse
|
11
|
Docosahexaenoic Acid Counteracts the Hypoxic-Induced Inflammatory and Metabolic Alterations in 3T3-L1 Adipocytes. Nutrients 2022; 14:nu14214600. [PMID: 36364860 PMCID: PMC9659308 DOI: 10.3390/nu14214600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/15/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
Background: Hypoxia is caused by the excessive expansion of the white adipose tissue (AT) and is associated with obesity-related conditions such as insulin resistance, inflammation, and oxidative stress. Docosahexaenoic acid (DHA) is an omega-3 fatty acid reported to have beneficial health effects. However, the effects of DHA in AT against hypoxia-induced immune-metabolic perturbations in adipocytes exposed to low O2 tension are not well known. Consequently, this study aimed to evaluate the impact of DHA on markers of inflammation, metabolism, apoptosis, and oxidative stress in 3T3-L1 cell adipocytes exposed to low O2 tension (1% O2) induced hypoxia. Methods: The apoptosis and reactive oxygen species (ROS) rates were evaluated. Metabolic parameters such as lactate, FFA, glycerol release, glucose uptake, and ATP content were assessed by a fluorometer. The expression of HIF-1, GLUT1 and the secretion of adipocytokines such as leptin, adiponectin, and pro-inflammatory markers was evaluated. Results: DHA-treated hypoxic cells showed significantly decreased basal free fatty acid release, lactate production, and enhanced glucose consumption. In addition, DHA-treatment of hypoxic cells caused a significant reduction in the apoptosis rate and ROS production with decreased lipid peroxidation. Moreover, DHA-treatment of hypoxic cells caused a decreased secretion of pro-inflammatory markers (IL-6, MCP-1) and leptin and increased adiponectin secretion compared with hypoxic cells. Furthermore, DHA-treatment of hypoxic cells caused significant reductions in the expression of genes related to hypoxia (HIF-1, HIF-2), anaerobic metabolism (GLUT1 and Ldha), ATP production (ANT2), and fat metabolism (FASN and PPARY). Conclusion: This study suggests that DHA can exert potential anti-obesity effects by reducing the secretion of inflammatory adipokines, oxidative stress, lipolysis, and apoptosis.
Collapse
|
12
|
Aria H, Rezaei M, Nazem S, Daraei A, Nikfar G, Mansoori B, Bahmanyar M, Tavassoli A, Vakil MK, Mansoori Y. Purinergic receptors are a key bottleneck in tumor metabolic reprogramming: The prime suspect in cancer therapeutic resistance. Front Immunol 2022; 13:947885. [PMID: 36072596 PMCID: PMC9444135 DOI: 10.3389/fimmu.2022.947885] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
ATP and other nucleoside phosphates have specific receptors named purinergic receptors. Purinergic receptors and ectonucleotidases regulate various signaling pathways that play a role in physiological and pathological processes. Extracellular ATP in the tumor microenvironment (TME) has a higher level than in normal tissues and plays a role in cancer cell growth, survival, angiogenesis, metastasis, and drug resistance. In this review, we investigated the role of purinergic receptors in the development of resistance to therapy through changes in tumor cell metabolism. When a cell transforms to neoplasia, its metabolic processes change. The metabolic reprogramming modified metabolic feature of the TME, that can cause impeding immune surveillance and promote cancer growth. The purinergic receptors contribute to therapy resistance by modifying cancer cells' glucose, lipid, and amino acid metabolism. Limiting the energy supply of cancer cells is one approach to overcoming resistance. Glycolysis inhibitors which reduce intracellular ATP levels may make cancer cells more susceptible to anti-cancer therapies. The loss of the P2X7R through glucose intolerance and decreased fatty acid metabolism reduces therapeutic resistance. Potential metabolic blockers that can be employed in combination with other therapies will aid in the discovery of new anti-cancer immunotherapy to overcome therapy resistance. Therefore, therapeutic interventions that are considered to inhibit cancer cell metabolism and purinergic receptors simultaneously can potentially reduce resistance to treatment.
Collapse
Affiliation(s)
- Hamid Aria
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marzieh Rezaei
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shima Nazem
- Department of Laboratory Medicine, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdolreza Daraei
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Ghasem Nikfar
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Behnam Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Maryam Bahmanyar
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Alireza Tavassoli
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Mohammad Kazem Vakil
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
13
|
Chitosan Oligosaccharides Alleviate H2O2-stimulated Granulosa Cell Damage via HIF-1α Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4247042. [PMID: 35401926 PMCID: PMC8993563 DOI: 10.1155/2022/4247042] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/02/2022] [Accepted: 03/02/2022] [Indexed: 12/23/2022]
Abstract
Oocyte maturation disorder and decreased quality are the main causes of infertility in women, and granulosa cells (GCs) provide the only microenvironment for oocyte maturation through autocrine and paracrine signaling by steroid hormones and growth factors. However, chronic inflammation and oxidative stress caused by ovarian hypoxia are the largest contributors to ovarian aging and GC dysfunction. Therefore, the amelioration of chronic inflammation and oxidative stress is expected to be a pivotal method to improve GC function and oocyte quality. In this study, we detected the protective effect of chitosan oligosaccharides (COS), on hydrogen peroxide- (H2O2-) stimulated oxidative damage in a human ovarian granulosa cell line (KGN). COS significantly increased cell viability, mitochondrial function, and the cellular glutathione (GSH) content and reduced apoptosis, reactive oxygen species (ROS) content, and the levels of 8-hydroxy-2′-deoxyguanosine (8-OHdG), 4-hydroxynonenal (4-HNE), hypoxia-inducible factor-1α (HIF-1α), and vascular endothelial-derived growth factor (VEGF) in H2O2-stimulated KGN cells. COS treatment significantly increased levels of the TGF-β1 and IL-10 proteins and decreased levels of the IL-6 protein. Compared with H2O2-stimulated KGN cells, COS significantly increased the levels of E2 and P4 and decreased SA-β-gal protein expression. Furthermore, COS caused significant inactivation of the HIF-1α-VEGF pathway in H2O2-stimulated KGN cells. Moreover, inhibition of this pathway enhanced the inhibitory effects of COS on H2O2-stimulated oxidative injury and apoptosis in GCs. Thus, COS protected GCs from H2O2-stimulated oxidative damage and apoptosis by inactivating the HIF-1α-VEGF signaling pathway. In the future, COS might represent a therapeutic approach for ameliorating disrupted follicle development.
Collapse
|
14
|
Chen SN, Xu ZG, Ma YX, Chen S, He GH, Han M, Gao X, Wang JH, Wu B, Wang J. Protective effect of LIF-huMSCs on the retina of diabetic model rats. Int J Ophthalmol 2021; 14:1508-1517. [PMID: 34667726 DOI: 10.18240/ijo.2021.10.06] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/07/2021] [Indexed: 12/26/2022] Open
Abstract
AIM To investigate the protective effect of human umbilical cord mesenchymal stem cells (hUCMSCs) modified by the LIF gene on the retinal function of diabetic model rats and preliminarily explore the possible mechanism. METHODS A stably transfected cell line of hUCMSCs overexpressing leukemia inhibitory factor (LIF) was constructed. Overexpression was verified by fluorescent quantitative polymerase chain reaction (qPCR). Forty-eight adult Sprague-Dawley rats were randomly divided into a normal control group (group A), streptozotocin-induced diabetic control group (group B), diabetic rats at 3mo injected with empty vector-transfected hUCMSCs (group C) or injected with LIF-hUCMSCs (group D). Four weeks after the intravitreal injection, analyses in all groups included retinal function using flash electroretinogram (F-ERG), retinal blood vessel examination of retinal flat mounts perfused with fluorescein isothiocyanate-dextran (FITC-dextran), and retinal structure examination of sections using hematoxylin and eosin staining. Expression levels of adiponectin (APN), high-sensitivity C-reactive protein (hs-CRP), and neurotrophin-4 (NT-4) in each group was detected using immunohistochemistry, PCR, Western blotting, and ELISA, respectively. RESULTS A stable transgenic cell line of LIF-hUCMSCs was constructed. F-ERG and FITC-dextran examinations revealed no abnormalities of retinal structure and function in group A, severe damage of the retinal blood vessels and function in group B, and improved retinal structure and function in group C and especially group D. qPCR, ELISA, and Western blot analyses revealed progressively higher APN and NT-4 expression levels in groups B, C, and D than in group A. hs-CRP expression was significantly higher in group B than in groups A, C, and D, and was significantly higher in group C than in group D (P<0.05). CONCLUSION LIF-hUCMSCs protect the retina of diabetic rats by upregulating APN and NT-4 expression and downregulating hs-CRP expression in the retina.
Collapse
Affiliation(s)
- Shan-Na Chen
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin 300070, China.,Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China.,Xiamen Kehong Eye Hospital, Xiamen 361000, Fujian Province, China
| | - Zhi-Gang Xu
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin 300070, China.,Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China.,Department of Ophthalmology, Baoan Central Hospital, Shenzhen 518000, China
| | - Ying-Xue Ma
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin 300070, China.,Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China.,Department of Ophthalmology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Song Chen
- Department of Vitreous and Retinopathy, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China
| | - Guang-Hui He
- Department of Vitreous and Retinopathy, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China
| | - Mei Han
- Department of Ophthalmology, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China
| | - Xiang Gao
- Studying for a Doctor of Medicine Degree from Nankai University, Tianjin 300000, China
| | - Jun-Hua Wang
- Department of Ophthalmology, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China
| | - Bin Wu
- Department of Ophthalmology, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China
| | - Jian Wang
- Department of Ophthalmology, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China
| |
Collapse
|
15
|
Al-Hejailan RS, Bakheet RH, Al-Saud MM, Al-Jufan MB, Al-Hindas HM, Al-Qattan SM, Al-Muhanna MK, Parhar RS, Conca W, Hansmann J, Collison KS, Walles H, Al-Mohanna FA. Toward allogenizing a xenograft: Xenogeneic cardiac scaffolds recellularized with human-induced pluripotent stem cells do not activate human naïve neutrophils. J Biomed Mater Res B Appl Biomater 2021; 110:691-701. [PMID: 34619017 DOI: 10.1002/jbm.b.34948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/31/2021] [Accepted: 09/22/2021] [Indexed: 11/12/2022]
Abstract
The limited availability of human donor organs suitable for transplantation has resulted in ever-increasing patient waiting lists globally. Xenotransplantation is considered a potential option, but is yet to reach clinical practice. Although remarkable progress has been made in overcoming immunological rejection, issues with functionality are still to be resolved. Bioengineering approaches have been used to create cardiac tissues with optimized functions. The use of decellularized xenogeneic cardiac tissues seeded with donor-derived cardiac cells may prove to be a viable strategy as supporting structures of the native tissue such as vasculature can be utilized. Here we used sequential perfusion to decellularize adult rat hearts. The acellular scaffolds were reseeded with human endothelial cells, human fibroblasts, human mesenchymal stem cells, and cardiac cells derived from human-induced pluripotent stem cells. The ability of the resultant recellularized rat scaffolds to activate human naïve neutrophils in vitro was investigated to measure xenogeneic recognition. Our results demonstrate that in contrast to cadaveric xenogeneic hearts, acellular and recellularized xenogeneic scaffolds did not activate human naïve neutrophils and suggest that decellularization removes the xenogeneic antigens that lead to human naïve neutrophil activation thus allowing human cells to populate the now "allogenized" xenogeneic scaffolds.
Collapse
Affiliation(s)
- Reem S Al-Hejailan
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Department of Heart Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Razan H Bakheet
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mashael M Al-Saud
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | - Hussain M Al-Hindas
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Somaya M Al-Qattan
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Muhanna K Al-Muhanna
- Tissue Engineering and Regenerative Medicine (TERM), Würzburg University, Würzburg, Germany
| | - Ranjit S Parhar
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Walter Conca
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Jan Hansmann
- Department of Heart Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Kate S Collison
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Heike Walles
- Department of Heart Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Futwan A Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
16
|
Redox Regulation of Lipid Mobilization in Adipose Tissues. Antioxidants (Basel) 2021; 10:antiox10071090. [PMID: 34356323 PMCID: PMC8301038 DOI: 10.3390/antiox10071090] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/14/2022] Open
Abstract
Lipid mobilization in adipose tissues, which includes lipogenesis and lipolysis, is a paramount process in regulating systemic energy metabolism. Reactive oxygen and nitrogen species (ROS and RNS) are byproducts of cellular metabolism that exert signaling functions in several cellular processes, including lipolysis and lipogenesis. During lipolysis, the adipose tissue generates ROS and RNS and thus requires a robust antioxidant response to maintain tight regulation of redox signaling. This review will discuss the production of ROS and RNS within the adipose tissue, their role in regulating lipolysis and lipogenesis, and the implications of antioxidants on lipid mobilization.
Collapse
|
17
|
Gao J, Zhu J, Zhao Y, Gan X, Yu H. Leptin attenuates hypoxia-induced apoptosis in human periodontal ligament cells via the reactive oxygen species-hypoxia-inducible factor-1α pathway. Exp Physiol 2021; 106:1752-1761. [PMID: 34143536 DOI: 10.1113/ep089324] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 06/17/2021] [Indexed: 02/05/2023]
Abstract
NEW FINDINGS What is the central question of this study? Does leptin have an effect on hypoxia-induced apoptosis in human periodontal ligament cells (hPDLCs), and what is the potential underlying mechanism? What is the main finding and its importance? Hypoxia induces cell apoptosis and leptin expression in hPDLCs through the induction of hypoxia-inducible factor-1α and accumulation of reactive oxygen species (ROS). Leptin shows feedback inhibition on hypoxia-induced ROS-mediated apoptosis in hPDLCs, suggesting a new application of leptin for hypoxic damage in periodontal diseases. ABSTRACT Hypoxia-induced apoptosis of human periodontal ligament cells (hPDLCs) is an important contributor to the progression of various periodontal diseases. Although leptin has been shown to protect connective tissue cells against hypoxia-induced injury, whether it might do so by attenuating hypoxia-induced apoptosis in hPDLCs remains unclear. Here, using CoCl2 treatment, we simulated hypoxic conditions in hPDLCs and explored whether apoptosis and reactive oxygen species (ROS) levels were related to hypoxia. After small interfering RNA (siRNA) inhibition of leptin and hypoxia-inducible factor-1α (HIF-1α), the levels of apoptosis, ROS and leptin expression were measured. We showed that in CoCl2 -treated hPDLCs, significantly higher cell apoptosis rates and ROS accumulation were observed. Cobalt chloride also increased leptin and HIF-1α expression in hPDLCs. Further investigation of the pathway demonstrated that inhibition of ROS attenuated hypoxia-induced cell apoptosis and leptin expression, whereas siRNA inhibition of leptin aggravated hypoxia-induced cell apoptosis and ROS accumulation. Hypoxia induces cell apoptosis and leptin expression in hPDLCs through the induction of ROS and HIF-1α pathways, and leptin shows feedback inhibition on ROS-mediated apoptosis in hPDLCs. These findings suggest a new application of leptin for hypoxic damage in periodontal diseases.
Collapse
Affiliation(s)
- Jing Gao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Junfei Zhu
- Stomatology Center, China Japan Friendship Hospital, Beijing, 100029, China
| | - Yuwei Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xueqi Gan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Haiyang Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
18
|
Fan J, Papadopoulos V. Mitochondrial TSPO Deficiency Triggers Retrograde Signaling in MA-10 Mouse Tumor Leydig Cells. Int J Mol Sci 2020; 22:ijms22010252. [PMID: 33383772 PMCID: PMC7795497 DOI: 10.3390/ijms22010252] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
The mitochondrial translocator protein (TSPO) has been shown to bind cholesterol with high affinity and is involved in mediating its availability for steroidogenesis. We recently reported that targeted Tspo gene deletion in MA-10 mouse tumor Leydig cells resulted in reduced cAMP-stimulated steroid formation and significant reduction in the mitochondrial membrane potential (ΔΨm) compared to control cells. We hypothesized that ΔΨm reduction in the absence of TSPO probably reflects the dysregulation and/or maintenance failure of some basic mitochondrial function(s). To explore the consequences of TSPO depletion via CRISPR-Cas9-mediated deletion (indel) mutation in MA-10 cells, we assessed the transcriptome changes in TSPO-mutant versus wild-type (Wt) cells using RNA-seq. Gene expression profiles were validated using real-time PCR. We report herein that there are significant changes in nuclear gene expression in Tspo mutant versus Wt cells. The identified transcriptome changes were mapped to several signaling pathways including the regulation of membrane potential, calcium signaling, extracellular matrix, and phagocytosis. This is a retrograde signaling pathway from the mitochondria to the nucleus and is probably the result of changes in expression of several transcription factors, including key members of the NF-κB pathway. In conclusion, TSPO regulates nuclear gene expression through intracellular signaling. This is the first evidence of a compensatory response to the loss of TSPO with transcriptome changes at the cellular level.
Collapse
Affiliation(s)
- Jinjiang Fan
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada;
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada;
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
- Correspondence: ; Fax: +1-323-442-1681
| |
Collapse
|
19
|
Jiang Z, Hsu JL, Li Y, Hortobagyi GN, Hung MC. Cancer Cell Metabolism Bolsters Immunotherapy Resistance by Promoting an Immunosuppressive Tumor Microenvironment. Front Oncol 2020; 10:1197. [PMID: 32775303 PMCID: PMC7387712 DOI: 10.3389/fonc.2020.01197] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/12/2020] [Indexed: 12/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting immune checkpoint proteins, such as CTLA-4 and PD-1/PD-L1, have demonstrated remarkable and durable clinical responses in various cancer types. However, a considerable number of patients receiving ICIs eventually experience a relapse due to diverse resistance mechanisms. As a result, there have been increasing research efforts to elucidate the molecular mechanisms behind resistance to ICIs and improve patient outcomes. There is growing evidence that the dysregulated metabolic activity of tumor cells generates an immunosuppressive tumor microenvironment (TME) that orchestrates an impaired anti-tumor immune response. Notably, the immunosuppressive TME is characterized by nutrient shortage, hypoxia, an acidic extracellular milieu, and abundant immunosuppressive molecules. A detailed understanding of the TME remains a major challenge in mounting a more effective anti-tumor immune response. Herein, we discuss how tumor cells reprogram metabolism to modulate a pro-tumor TME, driving disease progression and immune evasion; in particular, we highlight potential approaches to target metabolic vulnerabilities in the context of anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Zhou Jiang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jennifer L. Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yintao Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Gabriel N. Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Center for Molecular Medicine and Research Center for Cancer Biology, Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
20
|
CXCL13 is a differentiation- and hypoxia-induced adipocytokine that exacerbates the inflammatory phenotype of adipocytes through PHLPP1 induction. Biochem J 2020; 476:3533-3548. [PMID: 31710352 DOI: 10.1042/bcj20190709] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/04/2019] [Accepted: 11/11/2019] [Indexed: 01/16/2023]
Abstract
Hypoxia in adipose tissue is regarded as a trigger that induces dysregulation of the secretory profile in adipocytes. Similarly, local dysregulation of adipocytokine secretion is an initial event in the deleterious effects of obesity on metabolism. We previously reported that CXCL13 is highly produced during adipogenesis, however little is known about the roles of CXCL13 in adipocytes. Here, we found that hypoxia, as modeled by 1% O2 or exposure to the hypoxia-mimetic reagent desferrioxamine (DFO) has strong inductive effects on the expression of CXCL13 and CXCR5, a CXCL13 receptor, in both undifferentiated and differentiated adipocytes and in organ-cultured white adipose tissue (WAT). CXCL13 was also highly expressed in WAT from high fat diet-fed mice. Hypoxic profile, typified by increased expression of interleukin-6 (IL-6) and plasminogen activator inhibitor-1 (PAI-1) and decreased expression of adiponectin, was significantly induced by CXCL13 treatment during adipogenic differentiation. Conversely, the treatment of adipocytes with a neutralizing-antibody against CXCL13 as well as CXCR5 knockdown by specific siRNA effectively inhibited DFO-induced inflammation. The phosphorylation of Akt2, a protective factor of adipose inflammation, was significantly inhibited by CXCL13 treatment during adipogenic differentiation. Mechanistically, CXCL13 induces the expression of PHLPP1, an Akt2 phosphatase, through focal adhesion kinase (FAK) signaling; and correspondingly we show that CXCL13 and DFO-induced IL-6 and PAI-1 expression was blocked by Phlpp1 knockdown. Furthermore, we revealed the functional binding sites of PPARγ2 and HIF1-α within the Cxcl13 promoter. Taken together, these results indicate that CXCL13 is an adipocytokine that facilitates hypoxia-induced inflammation in adipocytes through FAK-mediated induction of PHLPP1 in autocrine and/or paracrine manner.
Collapse
|
21
|
Lin ZP, Lin HL, Yu XP, Zheng YJ, Cheng SY. TLR4 mediates inflammation and hepatic fibrosis induced by chronic intermittent hypoxia in rats. Mol Med Rep 2020; 22:651-660. [PMID: 32626927 PMCID: PMC7339543 DOI: 10.3892/mmr.2020.11134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/05/2020] [Indexed: 12/24/2022] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) is a common and complex disorder that is associated with liver injury. Moreover, previous studies have revealed that chronic intermittent hypoxia (CIH) is associated with the development of non-alcoholic fatty liver disease and hepatic fibrosis. However, the underlying molecular mechanisms remain largely unknown. The present study aimed to investigate whether chronic intermittent hypoxia induced hepatic fibrosis, in addition to determining its underlying mechanisms, in CIH model rats using immunohistochemistry, western blotting and reverse transcription-quantitative PCR. The present results suggested that CIH caused hepatic fibrosis and increased the expression levels of interleukin (IL)-1β, IL-8, monocyte chemotactic-1, tumor necrosis factor-α, intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 in the liver; these conditions could be reversed by Toll-like receptor 4 (TLR4) short hairpin RNA lentivirus treatment. Moreover, immunohistochemistry and western blotting results indicated that TLR4 and NF-κB expression levels were significantly increased in the CIH and CIH-TLR4 empty vector lentivirus group. However, protein expression levels of TLR4, NF-κB, inhibitor of NF-κB and phosphorylated-mitogen-activated protein kinase (MAPK)-1 in the hypoxia/reoxygenation group were significantly higher compared with the control group (P<0.05), and these results were reversed by the MAPK inhibitor U0126 in vitro. Collectively, the present preliminary results suggested that inflammation and the TLR4/NF-κB/MAPK signaling pathway may be involved in CIH-induced liver fibrosis.
Collapse
Affiliation(s)
- Zhi-Peng Lin
- Department of Infectious Diseases, The First Quanzhou Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Hui-Li Lin
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Xue-Ping Yu
- Department of Infectious Diseases, The First Quanzhou Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Yi-Juan Zheng
- Department of Infectious Diseases, The First Quanzhou Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Si-Yu Cheng
- Department of Infectious Diseases, The First Quanzhou Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| |
Collapse
|
22
|
Chen Y, Liu F, Han F, Lv L, Tang CE, Xie Z, Luo F. Omentin-1 is associated with atrial fibrillation in patients with cardiac valve disease. BMC Cardiovasc Disord 2020; 20:214. [PMID: 32375640 PMCID: PMC7203903 DOI: 10.1186/s12872-020-01478-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/12/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Epicardial adipose tissue (EAT) remodeling and adipocytokines are associated with structural remodeling in atrial fibrillation (AF). However, the role of omentin-1, a novel adipocytokine, in structural remodeling remains unknown. METHODS Hematoxylin and eosin (H&E) and Masson's trichrome stains were used to investigate the histology of EAT and right atrial appendages. The expression levels of adipocytokines in these human samples were determined by immunohistochemical assay and western blotting. Models of transforming growth factor (TGF)-β1-induced activation of cardiac fibroblasts (CFs) and TGF-β1-induced endothelial-mesenchymal transition (EndMT) of human umbilical vein endothelial cell (HUVEC) were established to explore roles of omentin-1 in these processes. To determine changes in adipocytokines secretion under hypoxia conditions, adipocytes were treated with 5% O2 and 95% N2, and then CFs and HUVECs were co-cultured with the conditioned medium of adipocytes to determine the effects of hypoxia-treated adipocytes on these cells. RESULTS Expression of omentin-1 was downregulated in the EAT and right atrial appendages from patients with AF compared to samples from patients without AF, while the TGF-β1 level was upregulated in EAT from patients with AF. EAT from patients with AF exhibited adipocyte hypertrophy and severe interstitial fibrosis. Omentin-1 inhibited TGF-β1-induced CF activation and reversed TGF-β1-induced HUVEC EndMT. Adipocytes treated with hypoxia exhibited downregulation of omentin-1 and partly activated CFs. CONCLUSIONS This study demonstrated that omentin-1 was an antifibrotic adipocytokine and was downregulated in patients with AF, which was partly mediated by hypoxia.
Collapse
Affiliation(s)
- Yubin Chen
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Fen Liu
- The Institute of Medical Science Research, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Fei Han
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lizhi Lv
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Can-E Tang
- The Institute of Medical Science Research, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhongshang Xie
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Fanyan Luo
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
23
|
Chronic Hypoxia-Induced Microvessel Proliferation and Basal Membrane Degradation in the Bone Marrow of Rats Regulated through the IL-6/JAK2/STAT3/MMP-9 Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9204708. [PMID: 32047820 PMCID: PMC7003287 DOI: 10.1155/2020/9204708] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 11/08/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022]
Abstract
Chronic hypoxia (CH) is characterized by long-term hypoxia that is associated with microvessel proliferation and basal membrane (BM) degradation in tissues. The IL-6/JAK2/STAT3/MMP-9 pathway has been described in a variety of human cancers and plays an essential role in microvessel proliferation and BM degradation. Therefore, this study investigated the role of the IL-6/JAK2/STAT3/MMP-9 pathway in hypoxia-mediated microvessel proliferation and BM degradation in the rat bone marrow. Eighty pathogen-free Sprague Dawley male rats were randomly divided into four groups (20 per group)—control group, CH group (exposed to hypoxia in a hypobaric chamber at a simulated altitude of 5000 m for 28 d), CH + STAT3 inhibitor group (7.5 mg/kg/d), and CH + DMSO group. Microvessel density (MVD) and BM degradation in the bone marrow were determined by immunofluorescence staining and transmission electron microscopy. Serum IL-6 levels were assessed by enzyme-linked immunosorbent assay (ELISA), and the levels of P-JAK2, P-STAT3, and MMP-9 were assessed by western blot analysis and real-time reverse transcription PCR (RT-PCR). Hypoxia increased serum IL-6 levels, which in turn increased JAK2 and STAT3 phosphorylation, which subsequently upregulated MMP-9. Overexpression of MMP-9 significantly promoted the elevation of MVD and BM degradation. Inhibition of STAT3 using an inhibitor, SH-4-54, significantly downregulated MMP-9 expression and decreased MVD and BM degradation. Surprisingly, STAT3 inhibition also decreased serum IL-6 levels and JAK2 phosphorylation. Our results suggest that the IL-6/JAK2/STAT3/MMP-9 pathway might be related to CH-induced microvessel proliferation and BM degradation in the bone marrow.
Collapse
|
24
|
Hong M, Shi H, Wang N, Tan HY, Wang Q, Feng Y. Dual Effects of Chinese Herbal Medicines on Angiogenesis in Cancer and Ischemic Stroke Treatments: Role of HIF-1 Network. Front Pharmacol 2019; 10:696. [PMID: 31297056 PMCID: PMC6606950 DOI: 10.3389/fphar.2019.00696] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1)-induced angiogenesis has been involved in numerous pathological conditions, and it may be harmful or beneficial depending on the types of diseases. Exploration on angiogenesis has sparked hopes in providing novel therapeutic approaches on multiple diseases with high mortality rates, such as cancer and ischemic stroke. The HIF-1 pathway is considered to be a major regulator of angiogenesis. HIF-1 seems to be involved in the vascular formation process by synergistic correlations with other proangiogenic factors in cancer and cerebrovascular disease. The regulation of HIF-1-dependent angiogenesis is related to the modulation of HIF-1 bioactivity by regulating HIF-1α transcription or protein translation, HIF-1α DNA binding, HIF-1α and HIF-1α dimerization, and HIF-1 degradation. Traditional Chinese herbal medicines have a long history of clinical use in both cancer and stroke treatments in Asia. Growing evidence has demonstrated potential proangiogenic benefits of Chinese herbal medicines in ischemic stroke, whereas tumor angiogenesis could be inhibited by the active components in Chinese herbal medicines. The objective of this review is to provide comprehensive insight on the effects of Chinese herbal medicines on angiogenesis by regulating HIF-1 pathways in both cancer and ischemic stroke.
Collapse
Affiliation(s)
- Ming Hong
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Honglian Shi
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, United States
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Hor-Yue Tan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| |
Collapse
|
25
|
Nader M, Alsolme E, Alotaibi S, Alsomali R, Bakheet D, Dzimiri N. SLMAP-3 is downregulated in human dilated ventricles and its overexpression promotes cardiomyocyte response to adrenergic stimuli by increasing intracellular calcium. Can J Physiol Pharmacol 2019; 97:623-630. [PMID: 30856349 DOI: 10.1139/cjpp-2018-0660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Structural dilation of cardiomyocytes (CMs) imposes a decline in cardiac performance that precipitates cardiac failure and sudden death. Since membrane proteins are implicated in dilated cardiomyopathy and heart failure, we evaluated the expression of the sarcolemmal membrane-associated protein (SLMAP) in dilated cardiomyopathy and its effect on CM contraction. We found that all 3 SLMAP isoforms (SLMAP-1, -2, and -3) are expressed in CMs and are downregulated in human dilated ventricles. Knockdown of SLMAPs in cultured CMs transduced with recombinant adeno-associated viral particles releasing SLMAP-shRNA precipitated reduced spontaneous contractile rate that was not fully recovered in SLMAP-depleted CMs challenged with isoproterenol (ISO), thus phenotypically mimicking heart failure performance. Interestingly, the overexpression of the SLMAP-3 full-length isoform induced a positive chronotropic effect in CMs that was more pronounced in response to ISO insult (vs. ISO-treated naïve CMs). Confocal live imaging showed that H9c2 cardiac myoblasts overexpressing SLMAP-3 exhibit a higher intracellular calcium transient peak when treated with ISO (vs. ISO-treated cells carrying a control adeno-associated viral particle). Proteomics revealed that SLMAP-3 interacts with the regulator of CM contraction, striatin. Collectively, our data demonstrate that SLMAP-3 is a novel regulator of CM contraction rate and their response to adrenergic stimuli. Loss of SLMAPs phenotypically mimics cardiac failure and crystallizes SLMAPs as predictive of dilated cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Moni Nader
- a Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia.,b Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Ebtehal Alsolme
- b Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Shahd Alotaibi
- b Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Rahmah Alsomali
- b Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Dana Bakheet
- b Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Nduna Dzimiri
- b Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
26
|
Wiecek M, Szymura J, Maciejczyk M, Kantorowicz M, Szygula Z. Acute Anaerobic Exercise Affects the Secretion of Asprosin, Irisin, and Other Cytokines - A Comparison Between Sexes. Front Physiol 2018; 9:1782. [PMID: 30618797 PMCID: PMC6295572 DOI: 10.3389/fphys.2018.01782] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/26/2018] [Indexed: 12/26/2022] Open
Abstract
Objective: The new adipokine, which is asprosin, affects glucose release from the liver to the blood, and thus, influences exercise metabolism. This is the first study assessing whether single anaerobic exercise affects asprosin secretion in women and men. Methods: 10 men and 10 women (aged 21.64 ± 1.22 and 22.64 ± 1.49, respectively) performed a single 20-s bicycle sprint. Blood samples were collected before exercise and in the 3′, 15′, 30′, and 60′ of recovery, and 24 h after competition. Results: Only in women did asprosin (P = 0.001) (15′, 30′, 60′, and 24 h after exercise) and irisin (P < 0.001) (15′, 30′, and 60′) concentrations increase. Leptin, however, decreased (P = 0.001) at 3′, 15′, and 30′ in women. There was an increase in interleukin-6 (P < 0.001) at 3′, 15′, 30′, and 60′ of recovery in men, at 15′, 30′, 60′, and 24 h of recovery in women, along with a simultaneous decrease in interleukin-1β (P < 0.001) at 15′, 30′, and 60′ of recovery in men, and at 15′ and 30′ of recovery in women (r = -0.35, P < 0.001). There was a positive correlation between asprosin and adiponectin and a negative one between asprosin and leptin. The increase in irisin concentration at 30′ of recovery was positively correlated with the increase in asprosin concentration and percentage fat content, while being negatively correlated with total and lean body mass (LBM). Conclusion: The single anaerobic effort induced an increase in asprosin and irisin secretion while reducing leptin secretion in women. Adipocytokine concentration changes are inter-related. Regardless of sex, anaerobic efforts induce anti-inflammatory effects.
Collapse
Affiliation(s)
- Magdalena Wiecek
- Department of Physiology and Biochemistry, Faculty of Physical Education and Sport, University of Physical Education in Krakow, Kraków, Poland
| | - Jadwiga Szymura
- Department of Clinical Rehabilitation, Faculty of Motor Rehabilitation, University of Physical Education in Krakow, Kraków, Poland
| | - Marcin Maciejczyk
- Department of Physiology and Biochemistry, Faculty of Physical Education and Sport, University of Physical Education in Krakow, Kraków, Poland
| | - Malgorzata Kantorowicz
- Faculty of Physical Education and Sport, University of Physical Education in Krakow, Kraków, Poland
| | - Zbigniew Szygula
- Department of Physiotherapy, State University of Applied Sciences in Nowy Sacz, Nowy Sącz, Poland
| |
Collapse
|
27
|
Takaya J, Yamanouchi S, Kino J, Tanabe Y, Kaneko K. A Calcium-Deficient Diet in Dams during Gestation Increases Insulin Resistance in Male Offspring. Nutrients 2018; 10:nu10111745. [PMID: 30428526 PMCID: PMC6266795 DOI: 10.3390/nu10111745] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 01/09/2023] Open
Abstract
Calcium (Ca) plays an important role in the pathogenesis of insulin resistance syndrome. Osteocalcin (OC), a bone formation biomarker, acts directly on β-cells and increases insulin secretion. We determined the effects of Ca deficiency during pregnancy and/or lactation on insulin resistance in offspring. Female Wistar rats consumed either a Ca-deficient or control diet ad libitum from three weeks preconception to 21 days postparturition. Pups were allowed to nurse their original mothers until weaning. The offspring were fed a control diet beginning at weaning and were killed on day 180. Serum carboxylated OC (Gla-OC) and undercarboxylated OC (Glu-OC), insulin and adipokines in offspring were measured. In males, mean levels of insulin, glucose, and HOMA-IR were higher in the Ca-deficient group than in the control group. In addition, ionized Ca (iCa) was inversely associated with serum Glu-OC and adiponectin in males. In females, mean levels of Glu-OC and Gla-OC in the Ca-deficient group were higher than in the control group. In all offspring, serum leptin levels were correlated with serum insulin levels, and inversely correlated with iCa. In conclusion, maternal Ca restriction during pregnancy and/or lactation influences postnatal offspring Ca metabolism and insulin resistance in a sex-specific manner.
Collapse
Affiliation(s)
- Junji Takaya
- Department of Pediatrics, Kansai Medical University, Hirakata 573-1010, Japan.
| | - Sohsaku Yamanouchi
- Department of Pediatrics, Kansai Medical University, Hirakata 573-1010, Japan.
| | - Jiro Kino
- Department of Pediatrics, Kansai Medical University, Hirakata 573-1010, Japan.
| | - Yuko Tanabe
- Department of Pediatrics, Kansai Medical University, Hirakata 573-1010, Japan.
| | - Kazunari Kaneko
- Department of Pediatrics, Kansai Medical University, Hirakata 573-1010, Japan.
| |
Collapse
|
28
|
Al-Anazi A, Parhar R, Saleh S, Al-Hijailan R, Inglis A, Al-Jufan M, Bazzi M, Hashmi S, Conca W, Collison K, Al-Mohanna F. Data on hypoxia-induced VEGF, leptin and NF-kB p65 expression. Data Brief 2018; 21:2395-2397. [PMID: 30547064 PMCID: PMC6282628 DOI: 10.1016/j.dib.2018.10.147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 10/24/2018] [Accepted: 10/27/2018] [Indexed: 12/18/2022] Open
Abstract
The data set presented here is associated with the article “Intracellular calcium and NF-kB regulate hypoxia-induced leptin, VEGF, IL-6 and adiponectin secretion in human adipocytes” (Al-Anazi et al., 2018). Data illustrate hypoxia-induced VEGF and leptin expression in human adipocytes treated with the calcium chelator BAPTA-AM (1 µM). It also shows NF-κB p65 induced expression by hypoxia. Preadipocytes were differentiated for 14 days and then subjected to 0.5–1.5% oxygen in the presence and absence of BAPTA-AM or the NF-κB inhibitor SN50 for 48 h prior to RNA isolation and PCR analysis.
Collapse
Affiliation(s)
| | - Ranjit Parhar
- Department of Cell Biology, Riyadh 11211, Saudi Arabia
| | - Soad Saleh
- Department of Cell Biology, Riyadh 11211, Saudi Arabia
| | | | - Angela Inglis
- Department of Cell Biology, Riyadh 11211, Saudi Arabia
| | - Mansour Al-Jufan
- Heart Centre, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Mohammed Bazzi
- Department of Biochemistry, College of Science, King Saud University, Riyadh 12372, Saudi Arabia
| | - Sarwar Hashmi
- Developmental Biology, Center for Vector Biology, Rutgers University, New Brunswick, NJ 08901, United States
| | - Walter Conca
- Department of Cell Biology, Riyadh 11211, Saudi Arabia.,Department of Medicine, King Faisal Specialist Hospital and Research Centre, Alfaisal University, Riyadh 11211, Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| | - Kate Collison
- Department of Cell Biology, Riyadh 11211, Saudi Arabia
| | - Futwan Al-Mohanna
- Department of Cell Biology, Riyadh 11211, Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| |
Collapse
|