1
|
Suryaningtyas IT, Marasinghe CK, Lee B, Je JY. Oral administration of PIISVYWK and FSVVPSPK peptides attenuates obesity, oxidative stress, and inflammation in high fat diet-induced obese mice. J Nutr Biochem 2025; 136:109791. [PMID: 39490639 DOI: 10.1016/j.jnutbio.2024.109791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/07/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
The bioactive peptides PIISVYWK (P1) and FSVVPSPK (P2), derived from the blue mussel Mytilus edulis, exhibit significant benefits in combating obesity, oxidative stress, and inflammation. This study demonstrates that these peptides inhibit the differentiation of bone marrow-derived mesenchymal stem cells (BMMSCs) into adipocytes by downregulating the adipogenic transcription factors peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT/enhancer-binding protein alpha (C/EBPα), and sterol regulatory element-binding protein 1 (SREBP-1). Furthermore, P1 and P2 reduce lipogenesis and enhance lipolysis through the activation of AMP-activated protein kinase (AMPK) and hormone-sensitive lipase (HSL). These peptides also decrease intracellular reactive oxygen species (ROS) generation during adipogenesis and inhibit the mitogen-activated protein kinase (MAPK) pathway, thereby reducing inflammation. The involvement of heme oxygenase-1 (HO-1) in this mechanism is confirmed by the reversal of these effects upon HO-1 inhibition. In vivo, oral administration of P1 and P2 in high-fat diet (HFD) obese mice prevents weight gain, reduces adipose tissue accumulation, lowers adipogenic and lipogenic biomarkers, improves serum cholesterol levels, enhances lipolysis, and decreases pro-inflammatory cytokine production. These findings suggest that P1 and P2 peptides may effectively prevent obesity and related metabolic disorders by activating the HO-1/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway.
Collapse
Affiliation(s)
- Indyaswan T Suryaningtyas
- Department of Food and Nutrition, Pukyong National University, Busan, Republic of Korea; Research Center for Food Technology and Processing, National Research and Innovation Agency, Yogyakarta, Indonesia
| | - Chathuri K Marasinghe
- Department of Food and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Bonggi Lee
- Department of Food and Nutrition, Pukyong National University, Busan, Republic of Korea
| | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan, Republic of Korea.
| |
Collapse
|
2
|
Di Stasi V, Contaldo A, Birtolo LI, Shahini E. Interplay of Cardiometabolic Syndrome and Biliary Tract Cancer: A Comprehensive Analysis with Gender-Specific Insights. Cancers (Basel) 2024; 16:3432. [PMID: 39410050 PMCID: PMC11476000 DOI: 10.3390/cancers16193432] [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: 08/24/2024] [Revised: 10/05/2024] [Accepted: 10/09/2024] [Indexed: 10/20/2024] Open
Abstract
BTC overall incidence is globally increasing. CCA, including its subtypes, is a form of BTC. MetS, obesity, MASLD, and diabetes are all linked to CCA in interconnected ways. The link between obesity and CCA is less well-defined in Eastern countries as compared to Western. Although more research is needed to determine the relationship between MASLD and extrahepatic CCA (eCCA), MASLD may be a concurrent risk factor for intrahepatic CCA, particularly in populations with established or unidentified underlying liver disease. Interestingly, the risk of biliary tract cancer (BTC) seemed to be higher in patients with shorter diabetes durations who were not treated with insulin. Therefore, early detection and prevention of chronic liver disease, as well as additional intervention studies, will undoubtedly be required to determine whether improvements to MetS, weight loss, and diabetes therapy can reduce the risk and progression of BTC. However, further studies are needed to understand how reproductive hormones are involved in causing BTC and to develop consistent treatment for patients. Finally, it is critical to carefully assess the cardiological risk in BTC patients due to their increased intrinsic cardiovascular risk, putting them at risk for thrombotic complications, cardiovascular death, cardiac metastasis, and nonbacterial thrombotic endocarditis. This review aimed to provide an updated summary of the relation between the abovementioned cardio-metabolic conditions and BTC.
Collapse
Affiliation(s)
- Vincenza Di Stasi
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| | - Antonella Contaldo
- Gastroenterology Unit, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| | - Lucia Ilaria Birtolo
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Umberto I Hospital, Sapienza University of Rome, 00185 Rome, Italy;
| | - Endrit Shahini
- Gastroenterology Unit, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| |
Collapse
|
3
|
Pérez-Gómez JM, Montero-Hidalgo AJ, Fuentes-Fayos AC, Sarmento-Cabral A, Guzmán-Ruiz R, Malagón MM, Herrera-Martínez AD, Gahete MD, Luque RM. Exploring the role of the inflammasomes on prostate cancer: Interplay with obesity. Rev Endocr Metab Disord 2023; 24:1165-1187. [PMID: 37819510 PMCID: PMC10697898 DOI: 10.1007/s11154-023-09838-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/21/2023] [Indexed: 10/13/2023]
Abstract
Obesity is a weight-related disorder characterized by excessive adipose tissue growth and dysfunction which leads to the onset of a systemic chronic low-grade inflammatory state. Likewise, inflammation is considered a classic cancer hallmark affecting several steps of carcinogenesis and tumor progression. In this regard, novel molecular complexes termed inflammasomes have been identified which are able to react to a wide spectrum of insults, impacting several metabolic-related disorders, but their contribution to cancer biology remains unclear. In this context, prostate cancer (PCa) has a markedly inflammatory component, and patients frequently are elderly individuals who exhibit weight-related disorders, being obesity the most prevalent condition. Therefore, inflammation, and specifically, inflammasome complexes, could be crucial players in the interplay between PCa and metabolic disorders. In this review, we will: 1) discuss the potential role of each inflammasome component (sensor, molecular adaptor, and targets) in PCa pathophysiology, placing special emphasis on IL-1β/NF-kB pathway and ROS and hypoxia influence; 2) explore the association between inflammasomes and obesity, and how these molecular complexes could act as the cornerstone between the obesity and PCa; and, 3) compile current clinical trials regarding inflammasome targeting, providing some insights about their potential use in the clinical practice.
Collapse
Affiliation(s)
- Jesús M Pérez-Gómez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Antonio J Montero-Hidalgo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - André Sarmento-Cabral
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Rocio Guzmán-Ruiz
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - María M Malagón
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Aura D Herrera-Martínez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Endocrinology and Nutrition Service, HURS/IMIBIC, Córdoba, Spain
| | - Manuel D Gahete
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain.
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain.
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain.
| |
Collapse
|
4
|
Azimnasab-Sorkhabi P, Soltani-Asl M, Kfoury JR, Algenstaedt P, Mehmetzade HF, Hashemi Aghdam Y. The impact of leptin and its receptor polymorphisms on type 1 diabetes in a population of northwest Iran. Ann Hum Biol 2022; 49:317-322. [PMID: 36218419 DOI: 10.1080/03014460.2022.2134453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
BACKGROUND Diabetes comprises a serious disease with significant growth in the number of cases in recent years. Here, we cover the gap in information between leptin (LEP) and type 1 diabetes in the Iranian population. AIM To recognise LEP G2548A and LEP receptor Q223R polymorphisms in Iranian people and their association with type 1 diabetes susceptibility. SUBJECTS AND METHODS Characteristics such as fasting blood sugar (FBS) were measured in 80 control non-diabetic individuals and 89 diabetic patients. Moreover, LEP G2548A and LEP receptor Q223R polymorphisms were genotyped using polymerase chain reaction-restriction fragment length polymorphism technique. RESULTS The frequency of the A allele was nearly three times greater in diabetes patients than in the control group. In addition, in the diabetes group, the AA genotype was five times greater than in the control group (p < 0.01). Furthermore, AA and AA + AG genotype models had higher FBS levels than the GG + AG and GG genotype models, respectively (p < 0.01). CONCLUSION The LEP G2548A polymorphism could be related to type 1 diabetes susceptibility, but not LEPR Q223R polymorphism in the Iranian population. Importantly, further studies are essential to examine the impact of LEP G2548A and LEPR Q223R polymorphisms in the endocrinology area.
Collapse
Affiliation(s)
- Parviz Azimnasab-Sorkhabi
- Department of Biology, Faculty of Science, Islamic Azad University, Iran.,Department of Surgery, School of Veterinary Medicine and Animal Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Maryam Soltani-Asl
- Department of Biology, Faculty of Science, Islamic Azad University, Iran.,Department of Surgery, School of Veterinary Medicine and Animal Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - José Roberto Kfoury
- Department of Surgery, School of Veterinary Medicine and Animal Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Petra Algenstaedt
- Medical Clinic and Polyclinic of University of Hamburg, Eppendorf University, Hamburg, Germany
| | | | - Yashar Hashemi Aghdam
- Department of Traumatology, Spine and Orthopedic Surgery, Asklepios Hospital Altona, Faculty of Medicine, University of Hamburg, Hamburg, Germany
| |
Collapse
|
5
|
Nehme R, Diab-Assaf M, Decombat C, Delort L, Caldefie-Chezet F. Targeting Adiponectin in Breast Cancer. Biomedicines 2022; 10:2958. [PMID: 36428526 PMCID: PMC9687473 DOI: 10.3390/biomedicines10112958] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Obesity and breast cancer are two major health issues that could be categorized as sincere threats to human health. In the last few decades, the relationship between obesity and cancer has been well established and extensively investigated. There is strong evidence that overweight and obesity increase the risk of postmenopausal breast cancer, and adipokines are the central players in this relationship. Produced and secreted predominantly by white adipose tissue, adiponectin is a bioactive molecule that exhibits numerous protective effects and is considered the guardian angel of adipokine. In the obesity-cancer relationship, more and more evidence shows that adiponectin may prevent and protect individuals from developing breast cancer. Recently, several updates have been published on the implication of adiponectin in regulating tumor development, progression, and metastases. In this review, we provide an updated overview of the metabolic signaling linking adiponectin and breast cancer in all its stages. On the other hand, we critically summarize all the available promising candidates that may reactivate these pathways mainly by targeting adiponectin receptors. These molecules could be synthetic small molecules or plant-based proteins. Interestingly, the advances in genomics have made it possible to create peptide sequences that could specifically replace human adiponectin, activate its receptor, and mimic its function. Thus, the obvious anti-cancer activity of adiponectin on breast cancer should be better exploited, and adiponectin must be regarded as a serious biomarker that should be targeted in order to confront this threatening disease.
Collapse
Affiliation(s)
- Rawan Nehme
- Université Clermont-Auvergne, INRAE, UNH Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Moléculaire et Pharmacologie Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Lebanon
| | - Caroline Decombat
- Université Clermont-Auvergne, INRAE, UNH Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| |
Collapse
|
6
|
Tran VTH, Pham DV, Choi DY, Park PH. Mitophagy Induction and Aryl Hydrocarbon Receptor-Mediated Redox Signaling Contribute to the Suppression of Breast Cancer Cell Growth by Taloxifene via Regulation of Inflammasomes Activation. Antioxid Redox Signal 2022; 37:1030-1050. [PMID: 35286219 DOI: 10.1089/ars.2021.0192] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aims: Raloxifene, a selective estrogen receptor (ER) modulator, has been reported to exert the tumor-suppressive effects in both ER-positive and ER-negative cancer cells; however, the mechanisms underlying its ER-independent anti-cancer effects are poorly understood. The NLRP3 inflammasome, a critical component of the innate immune system, has recently received growing attention owing to its multifaceted roles in various aspects of cancer development. The present study aimed at examining the involvement of NLRP3 inflammasomes in the anti-breast cancer effects of raloxifene and its underlying mechanisms. Results: Raloxifene significantly inhibited the activation of NLRP3 inflammasomes in various breast cancer cell lines. Importantly, forced expression of a gain-of-function variant of NLRP3 rescued breast cancer cells from growth arrest by raloxifene, suggesting that the suppression of NLRP3 inflammasomes activation mediates the raloxifene-induced inhibition of breast cancer growth. Mechanistically, raloxifene suppressed NLRP3 inflammasomes activation by lowering the cellular levels of reactive oxygen species (ROS) through the modulation of redox signaling mediated via aryl hydrocarbon receptor (AhR)-nuclear factor erythroid 2-related factor 2 (Nrf2)-heme oxygenase-1 (HO-1) axis or the impaired generation of mitochondrial ROS in a mitophagy-dependent manner. Further, the blockage of AhR signaling or inhibition of mitophagy abolished the tumor-suppressive effect of raloxifene in a human breast tumor xenograft model. Innovation: We elucidate a novel molecular mechanism underlying the breast tumor suppressing effect of raloxifene. Conclusion: The results observed in this study suggest that the modulation of NLRP3 inflammasomes activation is a critical event in the inhibition of breast tumor growth by raloxifene. Antioxid. Redox Signal. 37, 1030-1050.
Collapse
Affiliation(s)
- Van Thi-Hong Tran
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Duc-Vinh Pham
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Dong-Young Choi
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea
| |
Collapse
|
7
|
Zuo Y, Xiao T, Qiu X, Liu Z, Zhang S, Zhou N. Adiponectin reduces apoptosis of diabetic cardiomyocytes by regulating miR-711/TLR4 axis. Diabetol Metab Syndr 2022; 14:131. [PMID: 36114541 PMCID: PMC9479314 DOI: 10.1186/s13098-022-00904-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 09/02/2022] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To investigate the regulation of adiponectin/miR-711 on TLR4/NF-κB-mediated inflammatory response and diabetic cardiomyocyte apoptosis. METHODS Diabetes models were established using rats and H9c2 cardiomyocytes. qRT-PCR was used to detect adiponectin, miR-711, and TLR4. MTT, β-galactosidase staining, and flow cytometry were utilized to assess cell viability, senescence, and apoptosis, respectively. The colorimetric method was used to measure caspase-3 activity, DCFH-DA probes to detect ROS, and western blotting to determine the protein levels of Bax, Bcl-2, TLR4, and p-NF-κB p65. ELISA was performed to measure the levels of adiponectin, ICAM-1, MCP-1, and IL-1β. Dual-luciferase reporter system examined the targeting relationship between miR-711 and TLR4. H&E and TUNEL staining revealed myocardial structure and apoptosis, respectively. RESULTS Adiponectin and miR-711 were underexpressed and TLR4/NF-κB signaling pathway was activated in high glucose-treated H9c2 cells. High glucose treatment reduced viability, provoked inflammatory response, and accelerated senescence and apoptosis in H9c2 cells. miR-711 could bind TLR4 mRNA and inactivate TLR4/NF-κB signaling. Adiponectin treatment increased miR-711 expression and blocked TLR4/NF-κB signaling. Adiponectin/miR-711 reduced myocardial inflammation and apoptosis in diabetic rats. CONCLUSION Adiponectin inhibits inflammation and alleviates high glucose-induced cardiomyocyte apoptosis by blocking TLR4/NF-κB signaling pathway through miR-711.
Collapse
Affiliation(s)
- Yu Zuo
- Department of the Pre-Hospital First-Aid, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Tao Xiao
- Nursing Department, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, Hunan, 410013, People's Republic of China.
| | - Xiangdong Qiu
- Department of the Pre-Hospital First-Aid, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Zuoliang Liu
- Intensive Care Unit, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Shengnan Zhang
- Department of the Pre-Hospital First-Aid, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Na Zhou
- Department of Anesthesiology, Hunan Aerospace Hospital, Changsha, Hunan, 410205, People's Republic of China
| |
Collapse
|
8
|
Feng X, Xiao J, Bai L. Role of adiponectin in osteoarthritis. Front Cell Dev Biol 2022; 10:992764. [PMID: 36158216 PMCID: PMC9492855 DOI: 10.3389/fcell.2022.992764] [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: 07/13/2022] [Accepted: 08/17/2022] [Indexed: 11/20/2022] Open
Abstract
Osteoarthritis (OA) is a widespread and most common joint disease which leads to social cost increasing accompany with aging population. Surgery is often the final treatment option. The major progression of OA includes cartilage degradation caused by chondrocytes metabolism imbalance. So, the molecular mechanisms of action in chondrocytes may provide insights into treatment methods for OA. Adiponectin is an adipokine with many biological functions in the cell metabolism. Numerous studies have illustrated that adiponectin has diverse biological effects, such as inhibition of cell apoptosis. It regulates various functions in different organs, including muscle, adipose tissue, brain, and bone, and regulates skeletal homeostasis. However, the relationship between adiponectin and cell death in the progression of OA needs further investigation. We elaborate the structure and function and the effect of adiponectin and state the correlation and intersection between adiponectin, autophagy, inflammation, and OA. From the perspective of oxidative stress, apoptosis, pyroptosis, and autophagy, we discuss the possible association between adiponectin, chondrocyte metabolism, and inflammatory factor efforts in OA. What’s more, we summarize the possible treatment methods, including the use of adiponectin as a drug target, and highlight the potential future mechanistic research. In this review, we summarize the molecular pathways and mechanisms of action of adiponectin in chondrocyte inflammation and death and the pathogenesis of OA. We also review the research on adiponectin as a target for treating OA. These studies provide a novel perspective to explore more effective treatment options considering the complex interrelationship between inflammation and metabolism in OA.
Collapse
Affiliation(s)
- Xinyuan Feng
- Department of Orthopedic Surgery, Shengjing Hospital, China Medical University, Shenyang, China
| | - Jiaying Xiao
- Department of Internal Medicine Integrated Ward 2, Shengjing Hospital, China Medical University, Shenyang, China
| | - Lunhao Bai
- Department of Orthopedic Surgery, Shengjing Hospital, China Medical University, Shenyang, China
- *Correspondence: Lunhao Bai,
| |
Collapse
|
9
|
Lee S, Pham DV, Park PH. Sestrin2 induction contributes to anti-inflammatory responses and cell survival by globular adiponectin in macrophages. Arch Pharm Res 2021; 45:38-50. [PMID: 34797495 DOI: 10.1007/s12272-021-01364-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022]
Abstract
Adiponectin, an adipose tissue-derived hormone, exhibits a modulatory effect on cell death/survival and possesses potent anti-inflammatory properties. However, the underlying molecular mechanisms remain elusive. Sestrin2, a stress-inducible metabolic protein, has shown cytoprotective and inflammation-modulatory effects under stressful conditions. In this study, we examined the role of sestrin2 signaling in the modulation of cell survival and inflammatory responses by globular adiponectin (gAcrp) in macrophages. We observed that gAcrp induced a significant increase in sestrin2 expression in both RAW 264.7 murine macrophages and primary murine macrophages. Notably, gAcrp treatment markedly increased expression of hypoxia inducible factor-1 α (HIF-1α) and gene silencing of HIF-1α blocked sestrin2 induction by gAcrp. In addition, pretreatment with a pharmacological inhibitor of ERK or PI3K abrogated both sestrin2 and HIF-1α expression by gAcrp, indicating that ERK/PI3K-mediated HIF-1α signaling pathway plays a critical role in sestrin2 induction by gAcrp. Furthermore, sestrin2 induction is implicated in autophagy activation, and knockdown of sestrin2 prevented enhanced cell viability by gAcrp. Moreover, gene silencing of sestrin2 caused restoration of gAcrp-induced expression of anti-inflammatory genes in a gene-selective manner. Taken together, these results indicate that sestrin2 induction critically contributes to cell survival and anti-inflammatory responses by gAcrp in macrophages.
Collapse
Affiliation(s)
- Sumin Lee
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Duc-Vinh Pham
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea. .,Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea.
| |
Collapse
|
10
|
Raut PK, Lee HS, Joo SH, Chun KS. Thymoquinone induces oxidative stress-mediated apoptosis through downregulation of Jak2/STAT3 signaling pathway in human melanoma cells. Food Chem Toxicol 2021; 157:112604. [PMID: 34627931 DOI: 10.1016/j.fct.2021.112604] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 01/04/2023]
Abstract
Melanoma is a highly aggressive and treatment-resistant cancer, and the incidence and mortality rates are increasing worldwide. Thymoquinone (TQ) is the active component of Nigella sativa seed extracts and exerts anticancer effects in various cancer cells. However, the anticancer effects of TQ on melanoma and the underlying molecular mechanisms remain elusive. In this study, TQ treatment induced apoptosis in SK-MEL-28 cells. Interestingly, constitutive phosphorylation of Janus kinase 2 (Jak2) and signal transducer and activator of transcription 3 (STAT3) was markedly decreased following TQ treatment. Furthermore, TQ treatment downregulated STAT3-dependent genes including cyclin D1, D2, and D3 and survivin. Moreover, inhibition of Jak2/STAT3 using AG490, an inhibitor of Jak2 or genetic ablation of STAT3, abrogated the expression of target genes. TQ increased the levels of reactive oxygen species (ROS), whereas pretreatment with N-acetyl cysteine (NAC), a ROS scavenger, prevented the suppressive effect of TQ on Jak2/STAT3 activation and protected SK-MEL-28 cells from TQ-induced apoptosis. TQ administration further attenuated the growth of SK-MEL-28 tumor xenografts. Taken together, TQ induced apoptosis of SK-MEL-28 by hindering the Jak2/STAT3 signaling pathway through ROS generation. Our results support further development of TQ as a potential anticancer therapeutic agent for treating melanoma.
Collapse
Affiliation(s)
- Pawan Kumar Raut
- College of Pharmacy, Keimyung University, Daegu, 42601, South Korea
| | - Hui Seong Lee
- College of Pharmacy, Keimyung University, Daegu, 42601, South Korea
| | - Sang Hoon Joo
- College of Pharmacy, Daegu Catholic University, Gyeongbuk, 38430, South Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu, 42601, South Korea.
| |
Collapse
|
11
|
Saponaro C, Scarpi E, Sonnessa M, Cioffi A, Buccino F, Giotta F, Pastena MI, Zito FA, Mangia A. Prognostic Value of NLRP3 Inflammasome and TLR4 Expression in Breast Cancer Patients. Front Oncol 2021; 11:705331. [PMID: 34540671 PMCID: PMC8443770 DOI: 10.3389/fonc.2021.705331] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/17/2021] [Indexed: 01/13/2023] Open
Abstract
Inflammasome complexes play a pivotal role in different cancer types. NOD-like receptor protein 3 (NLRP3) inflammasome is one of the most well-studied inflammasomes. Activation of the NLRP3 inflammasome induces abnormal secretion of soluble cytokines, generating advantageous inflammatory surroundings that support tumor growth. The expression levels of the NLRP3, PYCARD and TLR4 were determined by immunohistochemistry in a cohort of primary invasive breast carcinomas (BCs). We observed different NLRP3 and PYCARD expressions in non-tumor vs tumor areas (p<0.0001). All the proteins were associated to more aggressive clinicopathological characteristics (tumor size, grade, tumor proliferative activity etc.). Univariate analyses were carried out and related Kaplan-Meier curves plotted for NLRP3, PYCARD and TLR4 expression. Patients with higher NLRP3 and TLR4 expression had worse 5-year disease-free survival (DFS) compared to patients with lower NLRP3 and TLR4 expression (p =0.021 and p = 0.009, respectively). In multivariate analysis, TLR4 was confirmed as independent prognostic factors for DFS (HR = 2.03, 95% CI 1.16–3.57, p = 0.014), and high NLRP3 expression showed a slight association with DFS (HR = 1.75, 95% CI 0.98–3.15, p = 0.06). In conclusion, we showed TLR4 expression as independent prognostic factors and we highlighted for the first time that high expression of NLRP3 is linked to a poor prognosis in BC patients. These results suggest that NLRP3 and TLR4 could be two new good prognostic factor for BC patients.
Collapse
Affiliation(s)
- Concetta Saponaro
- Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Emanuela Scarpi
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola (FC), Italy
| | - Margherita Sonnessa
- Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Antonella Cioffi
- Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Francesca Buccino
- Pathology Department, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Francesco Giotta
- Medical Oncology Unit, IRCCS-Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Maria Irene Pastena
- Pathology Department, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | | | - Anita Mangia
- Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| |
Collapse
|
12
|
Pu X, Chen D. Targeting Adipokines in Obesity-Related Tumors. Front Oncol 2021; 11:685923. [PMID: 34485124 PMCID: PMC8415167 DOI: 10.3389/fonc.2021.685923] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/22/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity, a global epidemic, is an independent risk factor for the occurrence and development of a variety of tumors, such as breast cancer, pancreatic cancer, ovarian cancer and colorectal cancer. Adipocytes are important endocrine cells in the tumor microenvironment of obesity-related tumors, which can secrete a variety of adipokines (such as leptin, adiponectin, estrogen, resistin, MIF and MCP-1, etc.), among which leptin, adiponectin and estrogen are the most in-depth and valuable ones. These adipokines are closely related to tumorigenesis and the progression of tumors. In recent years, more and more studies have shown that under chronic inflammatory conditions such as obesity, adipocytes secrete more adipokines to promote the tumorigenesis and development of tumors. However, it is worth noting that although adiponectin is also secreted by adipocytes, it has an anti-tumor effect, and can cross-talk with other adipokines (such as leptin and estrogen) and insulin to play an anti-tumor effect together. In addition, obesity is the main cause of insulin resistance, which can lead to the increase of the expression levels of insulin and insulin-like growth factor (IGF). As important regulators of blood glucose and lipid metabolism, insulin and IGF also play an important role in the progress of obesity related tumors. In view of the important role of adipokines secreted by adipocytes and insulin/IGF in tumors, this article not only elaborates leptin, adiponectin and estrogen secreted by adipocytes and their mechanism of action in the development of obesity- related tumors, but also introduces the relationship between insulin/IGF, a regulator of lipid metabolism, and obesity related tumors. At the same time, it briefly describes the cancer-promoting mechanism of resistin, MIF and MCP-1 in obesity-related tumors, and finally summarizes the specific treatment opinions and measures for various adipokines and insulin/insulin-like growth factors in recent years.
Collapse
Affiliation(s)
- Xi Pu
- Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Deyu Chen
- Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
13
|
Osataphan S, Mahankasuwan T, Saengboonmee C. Obesity and cholangiocarcinoma: A review of epidemiological and molecular associations. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2021; 28:1047-1059. [PMID: 34053180 DOI: 10.1002/jhbp.1001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/02/2021] [Accepted: 05/19/2021] [Indexed: 11/10/2022]
Abstract
Cholangiocarcinoma (CCA) is a malignancy of bile duct epithelium, and its incidence is increasing globally. Numerous factors are reported associated with an increased risk of CCA and vary among populations across different areas. Obesity is a major, worldwide public health problem that leads to several complications and is associated with increased cancer risk. Although several epidemiological studies have shown that obesity is likely associated with the increased risk of CCA, this association might be limited to Western countries. Multiple hormones, cytokines, and metabolite perturbations in obese states have been shown to enhance tumorigenicity and metastasis potentials. Understanding the biological linkage of obesity to CCA might lead to novel prevention and therapeutic approaches to CCA treatment. This review summarizes the current evidence and highlights the knowledge gaps regarding the relationship between obesity and CCA from epidemiological and molecular perspectives.
Collapse
Affiliation(s)
| | | | - Charupong Saengboonmee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
14
|
Faria SS, Costantini S, de Lima VCC, de Andrade VP, Rialland M, Cedric R, Budillon A, Magalhães KG. NLRP3 inflammasome-mediated cytokine production and pyroptosis cell death in breast cancer. J Biomed Sci 2021; 28:26. [PMID: 33840390 PMCID: PMC8040227 DOI: 10.1186/s12929-021-00724-8] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/03/2021] [Indexed: 12/27/2022] Open
Abstract
Breast cancer is the most diagnosed malignancy in women. Increasing evidence has highlighted the importance of chronic inflammation at the local and/or systemic level in breast cancer pathobiology, influencing its progression, metastatic potential and therapeutic outcome by altering the tumor immune microenvironment. These processes are mediated by a variety of cytokines, chemokines and growth factors that exert their biological functions either locally or distantly. Inflammasomes are protein signaling complexes that form in response to damage- and pathogen-associated molecular patterns (DAMPS and PAMPS), triggering the release of pro-inflammatory cytokines. The dysregulation of inflammasome activation can lead to the development of inflammatory diseases, neurodegeneration, and cancer. A crucial signaling pathway leading to acute and chronic inflammation occurs through the activation of NLRP3 inflammasome followed by caspase 1-dependent release of IL-1β and IL-18 pro-inflammatory cytokines, as well as, by gasdermin D-mediated pyroptotic cell death. In this review we focus on the role of NLRP3 inflammasome and its components in breast cancer signaling, highlighting that a more detailed understanding of the clinical relevance of these pathways could significantly contribute to the development of novel therapeutic strategies for breast cancer.
Collapse
Affiliation(s)
- Sara Socorro Faria
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, DF, Brazil
| | - Susan Costantini
- Experimental Pharmacology Unit - Laboratory of Mercogliano (AV), Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | | | | | - Mickaël Rialland
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1231, 21000, Dijon, France
- UFR Sciences de la Vie, Terre et Environnement, Université de Bourgogne Franche-Comté, 21000, Dijon, France
| | - Rebe Cedric
- Platform of Transfer in Cancer Biology, Centre Georges François Leclerc, 21000, Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1231, 21000, Dijon, France
| | - Alfredo Budillon
- Experimental Pharmacology Unit - Laboratory of Mercogliano (AV), Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, DF, Brazil.
| |
Collapse
|
15
|
Lin TC, Hsiao M. Leptin and Cancer: Updated Functional Roles in Carcinogenesis, Therapeutic Niches, and Developments. Int J Mol Sci 2021; 22:ijms22062870. [PMID: 33799880 PMCID: PMC8002181 DOI: 10.3390/ijms22062870] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Leptin is an obesity-associated adipokine that is known to regulate energy metabolism and reproduction and to control appetite via the leptin receptor. Recent work has identified specific cell types other than adipocytes that harbor leptin and leptin receptor expression, particularly in cancers and tumor microenvironments, and characterized the role of this signaling axis in cancer progression. Furthermore, the prognostic significance of leptin in various types of cancer and the ability to noninvasively detect leptin levels in serum samples have attracted attention for potential clinical applications. Emerging findings have demonstrated the direct and indirect biological effects of leptin in regulating cancer proliferation, metastasis, angiogenesis and chemoresistance, warranting the exploration of the underlying molecular mechanisms to develop a novel therapeutic strategy. In this review article, we summarize and integrate transcriptome and clinical data from cancer patients together with the recent findings related to the leptin signaling axis in the aforementioned malignant phenotypes. In addition, a comprehensive analysis of leptin and leptin receptor distribution in a pancancer panel and in individual cell types of specific organs at the single-cell level is presented, identifying those sites that are prone to leptin-mediated tumorigenesis. Our results shed light on the role of leptin in cancer and provide guidance and potential directions for further research for scientists in this field.
Collapse
Affiliation(s)
- Tsung-Chieh Lin
- Genomic Medicine Core Laboratory, Department of Medical Research and Development, Chang Gung Memorial Hospital, Linkou 333, Taiwan;
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-2-27871243; Fax: +886-2-27899931
| |
Collapse
|
16
|
Tumor Metabolic Reprogramming by Adipokines as a Critical Driver of Obesity-Associated Cancer Progression. Int J Mol Sci 2021; 22:ijms22031444. [PMID: 33535537 PMCID: PMC7867092 DOI: 10.3390/ijms22031444] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 12/11/2022] Open
Abstract
Adiposity is associated with an increased risk of various types of carcinoma. One of the plausible mechanisms underlying the tumor-promoting role of obesity is an aberrant secretion of adipokines, a group of hormones secreted from adipose tissue, which have exhibited both oncogenic and tumor-suppressing properties in an adipokine type- and context-dependent manner. Increasing evidence has indicated that these adipose tissue-derived hormones differentially modulate cancer cell-specific metabolism. Some adipokines, such as leptin, resistin, and visfatin, which are overproduced in obesity and widely implicated in different stages of cancer, promote cellular glucose and lipid metabolism. Conversely, adiponectin, an adipokine possessing potent anti-tumor activities, is linked to a more favorable metabolic phenotype. Adipokines may also play a pivotal role under the reciprocal regulation of metabolic rewiring of cancer cells in tumor microenvironment. Given the fact that metabolic reprogramming is one of the major hallmarks of cancer, understanding the modulatory effects of adipokines on alterations in cancer cell metabolism would provide insight into the crosstalk between obesity, adipokines, and tumorigenesis. In this review, we summarize recent insights into putative roles of adipokines as mediators of cellular metabolic rewiring in obesity-associated tumors, which plays a crucial role in determining the fate of tumor cells.
Collapse
|
17
|
Leptin, Leptin Receptor, KHDRBS1 (KH RNA Binding Domain Containing, Signal Transduction Associated 1), and Adiponectin in Bone Metastasis from Breast Carcinoma: An Immunohistochemical Study. Biomedicines 2020; 8:biomedicines8110510. [PMID: 33213024 PMCID: PMC7698510 DOI: 10.3390/biomedicines8110510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer patients are at a high risk of complications from bone metastasis. Molecular characterization of bone metastases is essential for the discovery of new therapeutic targets. Here, we investigated the expression and the intracellular distribution of KH RNA binding domain containing, signal transduction associated 1 (KHDRBS1), leptin, leptin receptor (LEPR), and adiponectin in bone metastasis from breast carcinoma and looked for correlations between the data. The expression of these proteins is known in breast carcinoma, but it has not been investigated in bone metastatic tissue to date. Immunohistochemical analysis was carried out on bone metastasis specimens, then semiquantitative evaluation of the results and the Pearson test were performed to determine eventual correlations. KHDRBS1 expression was significantly higher in the nuclei than in the cytosol of metastatic cells; LEPR was prevalently observed in the cytosol and the nuclei; leptin and adiponectin were found in metastatic cells and stromal cells; the strongest positive correlation was between nuclear KHDRBS1 and nuclear LEPR expression. Taken together, our findings support the importance of the leptin/LEPR/KHDRBS1 axis and of adiponectin in the progression of bone metastasis and suggest their potential application in pharmacological interventions.
Collapse
|
18
|
Recent insights on modulation of inflammasomes by adipokines: a critical event for the pathogenesis of obesity and metabolism-associated diseases. Arch Pharm Res 2020; 43:997-1016. [PMID: 33078304 DOI: 10.1007/s12272-020-01274-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/13/2020] [Indexed: 12/17/2022]
Abstract
Aberrant production of adipokines, a group of adipocytes-derived hormones, is considered one of the most important pathological characteristics of obesity. In individuals with obesity, beneficial adipokines, such as adiponectin are downregulated, whereas leptin and other pro-inflammatory adipokines are highly upregulated. Hence, the imbalance in levels of these adipokines is thought to promote the development of obesity-linked complications. However, the mechanisms by which adipokines contribute to the pathogenesis of various diseases have not been clearly understood. Inflammasomes represent key signaling platform that triggers the inflammatory and immune responses through the processing of the interleukin family of pro-inflammatory cytokines in a caspase-1-dependent manner. Beyond their traditional function as a component of the innate immune system, inflammasomes have been recently integrated into the pathological process of multiple metabolism- and obesity-related disorders such as cardiovascular diseases, diabetes, fatty liver disease, and cancer. Interestingly, emerging evidence also highlights the role of adipokines in the modulation of inflammasomes activation, making it a promising mechanism underlying distinct biological actions of adipokines in diseases driven by inflammation and metabolic disorders. In this review, we summarize the effects of adipokines, in particular adiponectin, leptin, visfatin and apelin, on inflammasomes activation and their implications in the pathophysiology of obesity-linked complications.
Collapse
|