The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium

Adipose tissue (AT) biology is linked to cardiovascular health since obesity is associated with cardiovascular disease (CVD) and positively correlated with excessive visceral fat accumulation. AT signaling to myocardial cells through soluble factors known as adipokines, cardiokines, branched-chain a...

Full description

Autores:
Bermúdez, Valmore
Durán, Pablo
Rojas, Edward
Díaz, María P.
Rivas, José
Nava, Manuel
Chací, Maricarmen
Cabrera de Bravo, Mayela
Carrasquero, Rubén
Cano Ponce, Clímaco
Górriz, José Luis
D'Marco, Luis
Tipo de recurso:
Fecha de publicación:
2021
Institución:
Universidad Simón Bolívar
Repositorio:
Repositorio Digital USB
Idioma:
eng
OAI Identifier:
oai:bonga.unisimon.edu.co:20.500.12442/8657
Acceso en línea:
https://hdl.handle.net/20.500.12442/8657
https://doi.org/10.3389/fendo.2021.735070
Palabra clave:
Adipose tissue
Myocardiocytes
Microbiota
Obesity
Inflammation
Rights
openAccess
License
Attribution-NonCommercial-NoDerivatives 4.0 Internacional
id USIMONBOL2_95f07ebd59cfe1f73f04debdacbab4dc
oai_identifier_str oai:bonga.unisimon.edu.co:20.500.12442/8657
network_acronym_str USIMONBOL2
network_name_str Repositorio Digital USB
repository_id_str
dc.title.eng.fl_str_mv The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium
title The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium
spellingShingle The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium
Adipose tissue
Myocardiocytes
Microbiota
Obesity
Inflammation
title_short The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium
title_full The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium
title_fullStr The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium
title_full_unstemmed The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium
title_sort The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium
dc.creator.fl_str_mv Bermúdez, Valmore
Durán, Pablo
Rojas, Edward
Díaz, María P.
Rivas, José
Nava, Manuel
Chací, Maricarmen
Cabrera de Bravo, Mayela
Carrasquero, Rubén
Cano Ponce, Clímaco
Górriz, José Luis
D'Marco, Luis
dc.contributor.author.none.fl_str_mv Bermúdez, Valmore
Durán, Pablo
Rojas, Edward
Díaz, María P.
Rivas, José
Nava, Manuel
Chací, Maricarmen
Cabrera de Bravo, Mayela
Carrasquero, Rubén
Cano Ponce, Clímaco
Górriz, José Luis
D'Marco, Luis
dc.subject.eng.fl_str_mv Adipose tissue
Myocardiocytes
Microbiota
Obesity
Inflammation
topic Adipose tissue
Myocardiocytes
Microbiota
Obesity
Inflammation
description Adipose tissue (AT) biology is linked to cardiovascular health since obesity is associated with cardiovascular disease (CVD) and positively correlated with excessive visceral fat accumulation. AT signaling to myocardial cells through soluble factors known as adipokines, cardiokines, branched-chain amino acids and small molecules like microRNAs, undoubtedly influence myocardial cells and AT function via the endocrine-paracrine mechanisms of action. Unfortunately, abnormal total and visceral adiposity can alter this harmonious signaling network, resulting in tissue hypoxia and monocyte/macrophage adipose infiltration occurring alongside expanded intra-abdominal and epicardial fat depots seen in the human obese phenotype. These processes promote an abnormal adipocyte proteomic reprogramming, whereby these cells become a source of abnormal signals, affecting vascular and myocardial tissues, leading to meta-inflammation, atrial fibrillation, coronary artery disease, heart hypertrophy, heart failure and myocardial infarction. This review first discusses the pathophysiology and consequences of adipose tissue expansion, particularly their association with meta-inflammation and microbiota dysbiosis. We also explore the precise mechanisms involved in metabolic reprogramming in AT that represent plausible causative factors for CVD. Finally, we clarify how lifestyle changes could promote improvement in myocardiocyte function in the context of changes in AT proteomics and a better gut microbiome profile to develop effective, non-pharmacologic approaches to CVD.
publishDate 2021
dc.date.accessioned.none.fl_str_mv 2021-10-06T15:17:08Z
dc.date.available.none.fl_str_mv 2021-10-06T15:17:08Z
dc.date.issued.none.fl_str_mv 2021
dc.type.coarversion.fl_str_mv http://purl.org/coar/version/c_970fb48d4fbd8a85
dc.type.coar.fl_str_mv http://purl.org/coar/resource_type/c_2df8fbb1
dc.type.driver.eng.fl_str_mv info:eu-repo/semantics/article
dc.type.spa.spa.fl_str_mv Artículo científico
dc.identifier.citation.eng.fl_str_mv Bermúdez V, Durán P, Rojas E, Díaz MP, Rivas J, Nava M, Chacín M, Cabrera de Bravo M, Carrasquero R, Ponce CC, Górriz JL and D´Marco L (2021) The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium. Front. Endocrinol. 12:735070. doi: 10.3389/fendo.2021.735070
dc.identifier.issn.none.fl_str_mv 16642392
dc.identifier.uri.none.fl_str_mv https://hdl.handle.net/20.500.12442/8657
dc.identifier.doi.none.fl_str_mv https://doi.org/10.3389/fendo.2021.735070
identifier_str_mv Bermúdez V, Durán P, Rojas E, Díaz MP, Rivas J, Nava M, Chacín M, Cabrera de Bravo M, Carrasquero R, Ponce CC, Górriz JL and D´Marco L (2021) The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium. Front. Endocrinol. 12:735070. doi: 10.3389/fendo.2021.735070
16642392
url https://hdl.handle.net/20.500.12442/8657
https://doi.org/10.3389/fendo.2021.735070
dc.language.iso.eng.fl_str_mv eng
language eng
dc.rights.*.fl_str_mv Attribution-NonCommercial-NoDerivatives 4.0 Internacional
dc.rights.coar.fl_str_mv http://purl.org/coar/access_right/c_abf2
dc.rights.uri.*.fl_str_mv http://creativecommons.org/licenses/by-nc-nd/4.0/
dc.rights.accessrights.eng.fl_str_mv info:eu-repo/semantics/openAccess
rights_invalid_str_mv Attribution-NonCommercial-NoDerivatives 4.0 Internacional
http://creativecommons.org/licenses/by-nc-nd/4.0/
http://purl.org/coar/access_right/c_abf2
eu_rights_str_mv openAccess
dc.format.mimetype.spa.fl_str_mv pdf
dc.publisher.eng.fl_str_mv Frontiers Media
dc.source.eng.fl_str_mv Frontiers in Endocrinology
dc.source.none.fl_str_mv Vol. 12 (2021)
institution Universidad Simón Bolívar
bitstream.url.fl_str_mv https://bonga.unisimon.edu.co/bitstreams/0f4257c3-18db-438b-b158-5f9c98298d90/download
https://bonga.unisimon.edu.co/bitstreams/b5967e31-bd03-43cf-bdc7-b2789bf55486/download
https://bonga.unisimon.edu.co/bitstreams/3c031e5e-d23b-43b8-8862-b15475786801/download
https://bonga.unisimon.edu.co/bitstreams/b1389a5a-1e9b-4f56-ba06-85a7c6fa1eee/download
https://bonga.unisimon.edu.co/bitstreams/98055058-2c13-4ac6-ad81-5bfd7d83a222/download
https://bonga.unisimon.edu.co/bitstreams/7dd81a2e-8d31-4335-9f56-e1ada6eb6518/download
https://bonga.unisimon.edu.co/bitstreams/fa36acde-3b7f-46dc-818c-c47cf5a7d427/download
bitstream.checksum.fl_str_mv 4460e5956bc1d1639be9ae6146a50347
733bec43a0bf5ade4d97db708e29b185
99ded17c699dd710b771919833abd341
763b57de281c64f954cdc0b386be58cc
d497161139e30a3de3dd9e26e116d5fc
ee248de9193d6c622d195731ebadd122
1934ac2cd8b3eb6ec920d98989665f28
bitstream.checksumAlgorithm.fl_str_mv MD5
MD5
MD5
MD5
MD5
MD5
MD5
repository.name.fl_str_mv Repositorio Digital Universidad Simón Bolívar
repository.mail.fl_str_mv repositorio.digital@unisimon.edu.co
_version_ 1812100533049098240
spelling Bermúdez, Valmore29f9aa18-16a4-4fd3-8ce5-ed94a0b8663aDurán, Pablo6bb3ca43-d725-4ff0-ae00-4eda6cdbeea4Rojas, Edward94d56218-0c78-40f2-a501-74feeca9191cDíaz, María P.e5017bb2-15b8-412f-a151-05cba49ffa99Rivas, Josée04c76cc-1f79-43a1-b7ad-ea11db9b8e23Nava, Manuelf9865b69-841a-4eea-b7da-6a174b033d10Chací, Maricarmend0dfd276-7638-450e-bee4-f11a5b30b2b8Cabrera de Bravo, Mayelad984e281-e460-420f-9c2a-4af12b674973Carrasquero, Rubén87b1437e-aa54-4ca0-ba7e-ccec38847d59Cano Ponce, Clímaco48f3cf35-b24c-4071-80c6-85776c27c0acGórriz, José Luis3bfbeb8b-bff2-4c54-92a6-8b35f29dccc6D'Marco, Luis4f289143-892b-43a3-ac1f-6f462224f3142021-10-06T15:17:08Z2021-10-06T15:17:08Z2021Bermúdez V, Durán P, Rojas E, Díaz MP, Rivas J, Nava M, Chacín M, Cabrera de Bravo M, Carrasquero R, Ponce CC, Górriz JL and D´Marco L (2021) The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium. Front. Endocrinol. 12:735070. doi: 10.3389/fendo.2021.73507016642392https://hdl.handle.net/20.500.12442/8657https://doi.org/10.3389/fendo.2021.735070Adipose tissue (AT) biology is linked to cardiovascular health since obesity is associated with cardiovascular disease (CVD) and positively correlated with excessive visceral fat accumulation. AT signaling to myocardial cells through soluble factors known as adipokines, cardiokines, branched-chain amino acids and small molecules like microRNAs, undoubtedly influence myocardial cells and AT function via the endocrine-paracrine mechanisms of action. Unfortunately, abnormal total and visceral adiposity can alter this harmonious signaling network, resulting in tissue hypoxia and monocyte/macrophage adipose infiltration occurring alongside expanded intra-abdominal and epicardial fat depots seen in the human obese phenotype. These processes promote an abnormal adipocyte proteomic reprogramming, whereby these cells become a source of abnormal signals, affecting vascular and myocardial tissues, leading to meta-inflammation, atrial fibrillation, coronary artery disease, heart hypertrophy, heart failure and myocardial infarction. This review first discusses the pathophysiology and consequences of adipose tissue expansion, particularly their association with meta-inflammation and microbiota dysbiosis. We also explore the precise mechanisms involved in metabolic reprogramming in AT that represent plausible causative factors for CVD. Finally, we clarify how lifestyle changes could promote improvement in myocardiocyte function in the context of changes in AT proteomics and a better gut microbiome profile to develop effective, non-pharmacologic approaches to CVD.pdfengFrontiers MediaAttribution-NonCommercial-NoDerivatives 4.0 Internacionalhttp://creativecommons.org/licenses/by-nc-nd/4.0/info:eu-repo/semantics/openAccesshttp://purl.org/coar/access_right/c_abf2Frontiers in EndocrinologyVol. 12 (2021)Adipose tissueMyocardiocytesMicrobiotaObesityInflammationThe Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardiuminfo:eu-repo/semantics/articleArtículo científicohttp://purl.org/coar/version/c_970fb48d4fbd8a85http://purl.org/coar/resource_type/c_2df8fbb1World Health Organization. Obesity and Overweight. Available at: https:// www.who.int/news-room/fact-sheets/detail/obesity-and-overweight (Accessed cited 2021 May 6).Cawley J, Meyerhoefer C. The Medical Care Costs of Obesity: An Instrumental Variables Approach. J Health Econ (2012) 31(1):219–30. doi: 10.1016/j.jhealeco.2011.10.003. Fruh SM. Obesity: Risk Factors, Complications, and Strategies for Sustainable Long-Term Weight Management. J Am Assoc Nurse Pract (2017) 29(S1):S3–14. doi: 10.1002/2327-6924.12510World Health Organization. Cardiovascular Diseases (CVDs). Available at: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases- (cvds) (Accessed cited 2021 May 6).Oikonomou EK, Antoniades C. The Role of Adipose Tissue in Cardiovascular Health and Disease. Nat Rev Cardiol (2019) 16(2):83–99. doi: 10.1038/s41569-018-0097-6Antoniades C. “Dysfunctional” Adipose Tissue in Cardiovascular Disease: A Reprogrammable Target or an Innocent Bystander? Cardiovasc Res (2017) 113(9):997–8. doi: 10.1093/cvr/cvx116Rodrıguez A, Becerril S, Ezquerro S, Me ́ ́ndez-Giménez L, Frühbeck G. Crosstalk Between Adipokines and Myokines in Fat Browning. Acta Physiol (Oxf) (2017) 219(2):362–81. doi: 10.1111/apha.12686Arner P, Kulyté A. MicroRNA Regulatory Networks in Human Adipose Tissue and Obesity. Nat Rev Endocrinol (2015) 11(5):276–88. doi: 10.1038/ nrendo.2015.25Ahima RS, Lazar MA. Adipokines and the Peripheral and Neural Control of Energy Balance. Mol Endocrinol (2008) 22(5):1023–31. doi: 10.1210/ me.2007-0529Bohan R, Tianyu X, Tiantian Z, Ruonan F, Hongtao H, Qiong W, et al. Gut Microbiota: A Potential Manipulator for Host Adipose Tissue and Energy Metabolism. J Nutr Biochem (2019) 64:206–17. doi: 10.1016/ j.jnutbio.2018.10.020. Matsuzawa Y, Shimomura I, Nakamura T, Keno Y, Kotani K, Tokunaga K. Pathophysiology and Pathogenesis of Visceral Fat Obesity. Obes Res (1995) 3(Suppl 2):187S–94S. doi: 10.1002/j.1550-8528.1995.tb00462.xVacca M, Di Eusanio M, Cariello M, Graziano G, D’Amore S, Petridis FD, et al. Integrative miRNA and Whole-Genome Analyses of Epicardial Adipose Tissue in Patients With Coronary Atherosclerosis. Cardiovasc Res (2016) 109(2):228–39. doi: 10.1093/cvr/cvv266Gao L, Mei S, Zhang S, Qin Q, Li H, Liao Y, et al. Cardio-Renal Exosomes in Myocardial Infarction Serum Regulate Proangiogenic Paracrine Signaling in Adipose Mesenchymal Stem Cells. Theranostics (2020) 10(3):1060–73. doi: 10.7150/thno.37678Tran K-V, Majka J, Sanghai S, Sardana M, Lessard D, Milstone Z, et al. Micro-RNAs Are Related to Epicardial Adipose Tissue in Participants With Atrial Fibrillation: Data From the MiRhythm Study. Front Cardiovasc Med (2019) 6:115. doi: 10.3389/fcvm.2019.00115Gan XT, Zhao G, Huang CX, Rowe AC, Purdham DM, Karmazyn M. Identification of Fat Mass and Obesity Associated (FTO) Protein Expression in Cardiomyocytes: Regulation by Leptin and Its Contribution to LeptinInduced Hypertrophy. PloS One (2013) 8(9):e74235. doi: 10.1371/journal. pone.0074235Hruby A, Hu FB. The Epidemiology of Obesity: A Big Picture. Pharmacoeconomics (2015) 33(7):673–89. doi: 10.1007/s40273-014-0243-xMoreira JBN, Wohlwend M, Wisløff U. Exercise and Cardiac Health: Physiological and Molecular Insights. Nat Metab (2020) 2(9):829–39. doi: 10.1038/s42255-020-0262-1Chait A, den Hartigh LJ. Adipose Tissue Distribution, Inflammation and Its Metabolic Consequences, Including Diabetes and Cardiovascular Disease. Front Cardiovasc Med (2020) 7:22. doi: 10.3389/fcvm.2020. 00022Cinti S. The Adipose Organ at a Glance. Dis Model Mech (2012) 5(5):588–94. doi: 10.1242/dmm.009662Mariman ECM, Wang P. Adipocyte Extracellular Matrix Composition, Dynamics and Role in Obesity. Cell Mol Life Sci (2010) 67(8):1277–92. doi: 10.1007/s00018-010-0263-4Badimon L, Cubedo J. Adipose Tissue Depots and Inflammation: Effects on Plasticity and Resident Mesenchymal Stem Cell Function. Cardiovasc Res (2017) 113(9):1064–73. doi: 10.1093/cvr/cvx096Romacho T, Elsen M, Röhrborn D, Eckel J. Adipose Tissue and Its Role in Organ Crosstalk. Acta Physiol (Oxf) (2014) 210(4):733–53. doi: 10.1111/ apha.12246Gaborit B, Venteclef N, Ancel P, Pelloux V, Gariboldi V, Leprince P, et al. Human Epicardial Adipose Tissue Has a Specific Transcriptomic Signature Depending on Its Anatomical Peri-Atrial, Peri-Ventricular, or PeriCoronary Location. Cardiovasc Res (2015) 108(1):62–73. doi: 10.1093/cvr/ cvv208Lanthier N, Leclercq IA. Adipose Tissues as Endocrine Target Organs. Best Pract Res Clin Gastroenterol (2014) 28(4):545–58. doi: 10.1016/j.bpg. 2014.07.002Kajimura S. Advances in the Understanding of Adipose Tissue Biology. Nat Rev Endocrinol (2017) 13(2):69–70. doi: 10.1038/nrendo.2016.211Iozzo P. Myocardial, Perivascular, and Epicardial Fat. Diabetes Care (2011) 34(Suppl 2):S371–9. doi: 10.2337/dc11-s250Ferrero KM, Koch WJ. Metabolic Crosstalk Between the Heart and Fat. Korean Circ J (2020) 50(5):379. doi: 10.4070/kcj.2019.0400Heeren J, Münzberg H. Novel Aspects of Brown Adipose Tissue Biology. Endocrinol Metab Clin North Am (2013) 42(1):89–107. doi: 10.1016/ j.ecl.2012.11.004Gaspar RC, Pauli JR, Shulman GI, Muñoz VR. An Update on Brown Adipose Tissue Biology: A Discussion of Recent Findings. Am J PhysiolEndocrinol Metab (2021) 320(3):E488–95. doi: 10.1152/ajpendo.00310.2020Saito M. Human Brown Adipose Tissue: Regulation and Anti-Obesity Potential Review. Endocr J (2014) 61(5):409–16. doi: 10.1507/ endocrj.EJ13-0527Leitner BP, Huang S, Brychta RJ, Duckworth CJ, Baskin AS, McGehee S, et al. Mapping of Human Brown Adipose Tissue in Lean and Obese Young Men. Proc Natl Acad Sci USA (2017) 114(32):8649–54. doi: 10.1073/ pnas.1705287114Frühbeck G, Becerril S, Sáinz N, Garrastachu P, Garcıa-Velloso MJ. BAT: A ́ New Target for Human Obesity? Trends Pharmacol Sci (2009) 30(8):387–96. doi: 10.1016/j.tips.2009.05.003Gaspar RC, Muñoz VR, Azevêdo Macêdo AP, Lins Vieira R, Pauli JR. A Palette of Adipose Tissue: Multiple Functionality and Extraordinary Plasticity. Trends Anat Physiol (2021) 4:013. doi: 10.24966/TAP-7752/ 100013Rosenwald M, Wolfrum C. The Origin and Definition of Brite Versus White and Classical Brown Adipocytes. Adipocyte (2014) 3(1):4–9. doi: 10.4161/ adip.26232Wu J, Cohen P, Spiegelman BM. Adaptive Thermogenesis in Adipocytes: Is Beige the New Brown? Genes Dev (2013) 27(3):234–50. doi: 10.1101/ gad.211649.112World Health Organization. Health Topics. Obesity (2021). Available at: https://www.who.int/health-topics/obesity#tab=tab_1.Rutkowski JM, Stern JH, Scherer PE. The Cell Biology of Fat Expansion. J Cell Biol (2015) 208(5):501–12. doi: 10.1083/jcb.201409063Ghaben AL, Scherer PE. Adipogenesis and Metabolic Health. Nat Rev Mol Cell Biol (2019) 20(4):242–58. doi: 10.1038/s41580-018-0093-zKusminski CM, Holland WL, Sun K, Park J, Spurgin SB, Lin Y, et al. MitoNEET-Driven Alterations in Adipocyte Mitochondrial Activity Reveal a Crucial Adaptive Process That Preserves Insulin Sensitivity in Obesity. Nat Med (2012) 18(10):1539–49. doi: 10.1038/nm.2899Xue R, Wan Y, Zhang S, Zhang Q, Ye H, Li Y. Role of Bone Morphogenetic Protein 4 in the Differentiation of Brown Fat-Like Adipocytes. Am J Physiol Endocrinol Metab (2014) 306(4):E363–372. doi: 10.1152/ajpendo.00119.2013Rosen ED, MacDougald OA. Adipocyte Differentiation From the Inside Out. Nat Rev Mol Cell Biol (2006) 7(12):885–96. doi: 10.1038/nrm2066Gustafson B, Hammarstedt A, Hedjazifar S, Smith U. Restricted Adipogenesis in Hypertrophic Obesity: The Role of WISP2, WNT, and BMP4. Diabetes (2013) 62(9):2997–3004. doi: 10.2337/db13-0473Karastergiou K, Mohamed-Ali V. The Autocrine and Paracrine Roles of Adipokines. Mol Cell Endocrinol (2010) 318(1–2):69–78. doi: 10.1016/ j.mce.2009.11.011Halberg N, Khan T, Trujillo ME, Wernstedt-Asterholm I, Attie AD, Sherwani S, et al. Hypoxia-Inducible Factor 1alpha Induces Fibrosis and Insulin Resistance in White Adipose Tissue. Mol Cell Biol (2009) 29 (16):4467–83. doi: 10.1128/MCB.00192-09Carrière A, Carmona M-C, Fernandez Y, Rigoulet M, Wenger RH, Pénicaud L, et al. Mitochondrial Reactive Oxygen Species Control the Transcription Factor CHOP-10/GADD153 and Adipocyte Differentiation: A Mechanism for Hypoxia-Dependent Effect. J Biol Chem (2004) 279(39):40462–9. doi: 10.1074/jbc.M407258200Trayhurn P. Hypoxia and Adipose Tissue Function and Dysfunction in Obesity. Physiol Rev (2013) 93(1):1–21. doi: 10.1152/physrev.00017.2012Wang B, Wood IS, Trayhurn P. PCR Arrays Identify Metallothionein-3 as a Highly Hypoxia-Inducible Gene in Human Adipocytes. Biochem Biophys Res Commun (2008) 368(1):88–93. doi: 10.1016/j.bbrc.2008.01.036Mazzatti D, Lim F-L, O’Hara A, Wood IS, Trayhurn P. A Microarray Analysis of the Hypoxia-Induced Modulation of Gene Expression in Human Adipocytes. Arch Physiol Biochem (2012) 118(3):112–20. doi: 10.3109/ 13813455.2012.654611Engin A. Adipose Tissue Hypoxia in Obesity and Its Impact on Preadipocytes and Macrophages: Hypoxia Hypothesis. Adv Exp Med Biol (2017) 960:305–26. doi: 10.1007/978-3-319-48382-5_13Cildir G, Akıncılar SC, Tergaonkar V. Chronic Adipose Tissue Inflammation: All Immune Cells on the Stage. Trends Mol Med (2013) 19 (8):487–500. doi: 10.1016/j.molmed.2013.05.001Guzik TJ, Skiba DS, Touyz RM, Harrison DG. The Role of Infiltrating Immune Cells in Dysfunctional Adipose Tissue. Cardiovasc Res (2017) 113 (9):1009–23. doi: 10.1093/cvr/cvx108Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW. Obesity Is Associated With Macrophage Accumulation in Adipose Tissue. J Clin Invest (2003) 112(12):1796–808. doi: 10.1172/JCI200319246Amano SU, Cohen JL, Vangala P, Tencerova M, Nicoloro SM, Yawe JC, et al. Local Proliferation of Macrophages Contributes to Obesity-Associated Adipose Tissue Inflammation. Cell Metab (2014) 19(1):162–71. doi: 10.1016/j.cmet.2013.11.017Fujisaka S, Usui I, Bukhari A, Ikutani M, Oya T, Kanatani Y, et al. Regulatory Mechanisms for Adipose Tissue M1 and M2 Macrophages in Diet-Induced Obese Mice. Diabetes (2009) 58(11):2574–82. doi: 10.2337/db08-1475Kratz M, Coats BR, Hisert KB, Hagman D, Mutskov V, Peris E, et al. Metabolic Dysfunction Drives a Mechanistically Distinct Pro-Inflammatory Phenotype in Adipose Tissue Macrophages. Cell Metab (2014) 20(4):614–25. doi: 10.1016/j.cmet.2014.08.010Patsouris D, Li P-P, Thapar D, Chapman J, Olefsky JM, Neels JG. Ablation of CD11c-Positive Cells Normalises Insulin Sensitivity in Obese Insulin Resistant Animals. Cell Metab (2008) 8(4):301–9. doi: 10.1016/j.cmet. 2008.08.015Sun K, Kusminski CM, Scherer PE. Adipose Tissue Remodeling and Obesity. J Clin Invest (2011) 121(6):2094–101. doi: 10.1172/JCI45887Cinti S, Mitchell G, Barbatelli G, Murano I, Ceresi E, Faloia E, et al. Adipocyte Death Defines Macrophage Localisation and Function in Adipose Tissue of Obese Mice and Humans. J Lipid Res (2005) 46 (11):2347–55. doi: 10.1194/jlr.M500294-JLR200Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, et al. CD8+ Effector T Cells Contribute to Macrophage Recruitment and Adipose Tissue Inflammation in Obesity. Nat Med (2009) 15(8):914–20. doi: 10.1038/ nm.1964Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, et al. B Cells Promote Insulin Resistance Through Modulation of T Cells and Production of Pathogenic IgG Antibodies. Nat Med (2011) 17(5):610–7. doi: 10.1038/ nm.2353Mansuy-Aubert V, Zhou QL, Xie X, Gong Z, Huang J-Y, Khan AR, et al. Imbalance Between Neutrophil Elastase and Its Inhibitor a1-Antitrypsin in Obesity Alters Insulin Sensitivity, Inflammation, and Energy Expenditure. Cell Metab (2013) 17(4):534–48. doi: 10.1016/j.cmet.2013.03.005Talukdar S, Oh DY, Bandyopadhyay G, Li D, Xu J, McNelis J, et al. Neutrophils Mediate Insulin Resistance in Mice Fed a High-Fat Diet Through Secreted Elastase. Nat Med (2012) 18(9):1407–12. doi: 10.1038/ nm.2885Pasarica M, Sereda OR, Redman LM, Albarado DC, Hymel DT, Roan LE, et al. Reduced Adipose Tissue Oxygenation in Human Obesity: Evidence for Rarefaction, Macrophage Chemotaxis, and Inflammation Without an Angiogenic Response. Diabetes (2009) 58(3):718–25. doi: 10.2337/db08- 1098Sun K, Wernstedt Asterholm I, Kusminski CM, Bueno AC, Wang ZV, Pollard JW, et al. Dichotomous Effects of VEGF-A on Adipose Tissue Dysfunction. Proc Natl Acad Sci USA (2012) 109(15):5874–9. doi: 10.1073/ pnas.1200447109Villaret A, Galitzky J, Decaunes P, Estève D, Marques M-A, Sengenès C, et al. Adipose Tissue Endothelial Cells From Obese Human Subjects: Differences Among Depots in Angiogenic, Metabolic, and Inflammatory Gene Expression and Cellular Senescence. Diabetes (2010) 59(11):2755–63. doi: 10.2337/db10-0398Kikuchi R, Nakamura K, MacLauchlan S, Ngo DT-M, Shimizu I, Fuster JJ, et al. An Anti-Angiogenic Isoform of VEGF-A Contributes to Impaired Vascularization in Peripheral Artery Disease. Nat Med (2014) 20(12):1464– 71. doi: 10.1038/nm.3703Nishimura S, Manabe I, Nagasaki M, Seo K, Yamashita H, Hosoya Y, et al. In Vivo Imaging in Mice Reveals Local Cell Dynamics and Inflammation in Obese Adipose Tissue. J Clin Invest (2008) 118(2):710–21. doi: 10.1172/ JCI33328Greenstein AS, Khavandi K, Withers SB, Sonoyama K, Clancy O, Jeziorska M, et al. Local Inflammation and Hypoxia Abolish the Protective Anticontractile Properties of Perivascular Fat in Obese Patients. Circulation (2009) 119 (12):1661–70. doi: 10.1161/CIRCULATIONAHA.108.821181Berg G, Miksztowicz V, Morales C, Barchuk M. Epicardial Adipose Tissue in Cardiovascular Disease. Adv Exp Med Biol (2019) 1127:131–43. doi: 10.1007/ 978-3-030-11488-6_9. Spencer M, Unal R, Zhu B, Rasouli N, McGehee RE, Peterson CA, et al. Adipose Tissue Extracellular Matrix and Vascular Abnormalities in Obesity and Insulin Resistance. J Clin Endocrinol Metab (2011) 96(12):E1990–8. doi: 10.1210/jc.2011-1567Sun K, Park J, Gupta OT, Holland WL, Auerbach P, Zhang N, et al. Endotrophin Triggers Adipose Tissue Fibrosis and Metabolic Dysfunction. Nat Commun (2014) 5:3485. doi: 10.1038/ncomms4485Khan T, Muise ES, Iyengar P, Wang ZV, Chandalia M, Abate N, et al. Metabolic Dysregulation and Adipose Tissue Fibrosis: Role of Collagen VI. Mol Cell Biol (2009) 29(6):1575–91. doi: 10.1128/MCB.01300-08Geiger K, Leiherer A, Muendlein A, Stark N, Geller-Rhomberg S, Saely CH, et al. Identification of Hypoxia-Induced Genes in Human SGBS Adipocytes by Microarray Analysis. PloS One (2011) 6(10):e26465. doi: 10.1371/ journal.pone.0026465Yin J, Gao Z, He Q, Zhou D, Guo Z, Ye J. Role of Hypoxia in ObesityInduced Disorders of Glucose and Lipid Metabolism in Adipose Tissue. Am J Physiol Endocrinol Metab (2009) 296(2):E333–42. doi: 10.1152/ ajpendo.90760.2008Hayashi M, Sakata M, Takeda T, Yamamoto T, Okamoto Y, Sawada K, et al. Induction of Glucose Transporter 1 Expression Through HypoxiaInducible Factor 1alpha Under Hypoxic Conditions in TrophoblastDerived Cells. J Endocrinol (2004) 183(1):145–54. doi: 10.1677/joe. 1.05599Park HS, Kim JH, Sun BK, Song SU, Suh W, Sung J-H. Hypoxia Induces Glucose Uptake and Metabolism of Adipose−Derived Stem Cells. Mol Med Rep (2016) 14(5):4706–14. doi: 10.3892/mmr.2016.5796Mylonis I, Simos G, Paraskeva E. Hypoxia-Inducible Factors and the Regulation of Lipid Metabolism. Cells (2019) 8(3):214. doi: 10.3390/ cells8030214Barchetta I, Cimini FA, Ciccarelli G, Baroni MG, Cavallo MG. Sick Fat: The Good and the Bad of Old and New Circulating Markers of Adipose Tissue Inflammation. J Endocrinol Invest (2019) 42(11):1257–72. doi: 10.1007/ s40618-019-01052-3Chakrabarti P, Kim JY, Singh M, Shin Y-K, Kim J, Kumbrink J, et al. Insulin Inhibits Lipolysis in Adipocytes via the Evolutionarily Conserved Mtorc1- Egr1-ATGL-Mediated Pathway. Mol Cell Biol (2013) 33(18):3659–66. doi: 10.1128/MCB.01584-12Arner P, Rydén M. Fatty Acids, Obesity and Insulin Resistance. Obes Facts (2015) 8(2):147–55. doi: 10.1159/000381224Böni-Schnetzler M, Boller S, Debray S, Bouzakri K, Meier DT, Prazak R, et al. Free Fatty Acids Induce a Pro-Inflammatory Response in Islets via the Abundantly Expressed Interleukin-1 Receptor I. Endocrinology (2009) 150 (12):5218–29. doi: 10.1210/en.2009-0543Karastergiou K, Fried SK. Multiple Adipose Depots Increase Cardiovascular Risk via Local and Systemic Effects. Curr Atheroscler Rep (2013) 15(10):361. doi: 10.1007/s11883-013-0361-5Villarroya F, Cereijo R, Gavaldà-Navarro A, Villarroya J, Giralt M. Inflammation of Brown/Beige Adipose Tissues in Obesity and Metabolic Disease. J Intern Med (2018) 284(5):492–504. doi: 10.1111/joim.12803Ferré P, Burnol AF, Leturque A, Terretaz J, Penicaud L, Jeanrenaud B, et al. Glucose Utilisation In Vivo and Insulin-Sensitivity of Rat Brown Adipose Tissue in Various Physiological and Pathological Conditions. Biochem J (1986) 233(1):249–52. doi: 10.1042/bj2330249Orava J, Nuutila P, Noponen T, Parkkola R, Viljanen T, Enerbäck S, et al. Blunted Metabolic Responses to Cold and Insulin Stimulation in Brown Adipose Tissue of Obese Humans. Obes (Silver Spring) (2013) 21(11):2279– 87. doi: 10.1002/oby.20456. Estève D, Boulet N, Volat F, Zakaroff-Girard A, Ledoux S, Coupaye M, et al. Human White and Brite Adipogenesis Is Supported by MSCA1 and Is Impaired by Immune Cells. Stem Cells (2015) 33(4):1277–91. doi: 10.1002/ stem.1916Hills RD, Pontefract BA, Mishcon HR, Black CA, Sutton SC, Theberge CR. Gut Microbiome: Profound Implications for Diet and Disease. Nutrients (2019) 11(7):1613. doi: 10.3390/nu11071613Rampelli S, Guenther K, Turroni S, Wolters M, Veidebaum T, Kourides Y, et al. Pre-Obese Children’s Dysbiotic Gut Microbiome and Unhealthy Diets may Predict the Development of Obesity. Commun Biol (2018) 1(1):222. doi: 10.1155/2016/1629236Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, et al. The Gut Microbiota as an Environmental Factor That Regulates Fat Storage. Proc Natl Acad Sci (2004) 101(44):15718–23. doi: 10.1073/pnas.0407076101Bäckhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms Underlying the Resistance to Diet-Induced Obesity in Germ-Free Mice. Proc Natl Acad Sci USA (2007) 104(3):979–84. doi: 10.1073/pnas. 0605374104Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, et al. Gut Microbiota From Twins Discordant for Obesity Modulate Metabolism in Mice. Science (2013) 341(6150):1241214. doi: 10.1126/science.1241214Ghosh SS, Wang J, Yannie PJ, Ghosh S. Intestinal Barrier Dysfunction, LPS Translocation, and Disease Development. J Endocr Soc (2020) 4(2):bvz039. doi: 10.1210/jendso/bvz039Rohr MW, Narasimhulu CA, Rudeski-Rohr TA, Parthasarathy S. Negative Effects of a High-Fat Diet on Intestinal Permeability: A Review. Adv Nutr (2020) 11(1):77–91. doi: 10.1093/advances/nmz061Guo S, Al-Sadi R, Said HM, Ma TY. Lipopolysaccharide Causes an Increase in Intestinal Tight Junction Permeability In Vitro and In Vivo by Inducing Enterocyte Membrane Expression and Localisation of TLR-4 and CD14. Am J Pathol (2013) 182(2):375–87. doi: 10.1016/j.ajpath.2012.10.014Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic Endotoxemia Initiates Obesity and Insulin Resistance. Diabetes (2007) 56(7):1761–72. doi: 10.2337/db06-1491Song MJ, Kim KH, Yoon JM, Kim JB. Activation of Toll-Like Receptor 4 Is Associated With Insulin Resistance in Adipocytes. Biochem Biophys Res Commun (2006) 346(3):739–45. doi: 10.1016/j.bbrc.2006.05.170Caesar R, Tremaroli V, Kovatcheva-Datchary P, Cani PD, Bäckhed F. Crosstalk Between Gut Microbiota and Dietary Lipids Aggravates WAT Inflammation Through TLR Signaling. Cell Metab (2015) 22(4):658–68. doi: 10.1016/j.cmet.2015.07.026Cao W, Huang H, Xia T, Liu C, Muhammad S, Sun C. Homeobox A5 Promotes White Adipose Tissue Browning Through Inhibition of the Tenascin C/Toll-Like Receptor 4/Nuclear Factor Kappa B Inflammatory Signaling in Mice. Front Immunol (2018) 9:647. doi: 10.3389/fimmu.2018.00647Okla M, Wang W, Kang I, Pashaj A, Carr T, Chung S. Activation of Toll-Like Receptor 4 (TLR4) Attenuates Adaptive Thermogenesis via Endoplasmic Reticulum Stress. J Biol Chem (2015) 290(44):26476–90. doi: 10.1074/ jbc.M115.677724Muccioli GG, Naslain D, Bäckhed F, Reigstad CS, Lambert DM, Delzenne NM, et al. The Endocannabinoid System Links Gut Microbiota to Adipogenesis. Mol Syst Biol (2010) 6:392. doi: 10.1038/msb.2010.46Myhrstad MCW, Tunsjø H, Charnock C, Telle-Hansen VH. Dietary Fiber, Gut Microbiota, and Metabolic Regulation-Current Status in Human Randomized Trials. Nutrients (2020) 12(3):859. doi: 10.3390/nu12030859Blachier F, Mariotti F, Huneau JF, Tomé D. Effects of Amino Acid-Derived Luminal Metabolites on the Colonic Epithelium and Physiopathological Consequences. Amino Acids (2007) 33(4):547–62. doi: 10.1007/s00726-006- 0477-9Kim CH. Microbiota or Short-Chain Fatty Acids: Which Regulates Diabetes? Cell Mol Immunol (2018) 15(2):88–91. doi: 10.1038/cmi.2017.57Postler TS, Ghosh S. Understanding the Holobiont: How Microbial Metabolites Affect Human Health and Shape the Immune System. Cell Metab (2017) 26(1):110–30. doi: 10.1016/j.cmet.2017.05.008Ratajczak W, Rył A, Mizerski A, Walczakiewicz K, Sipak O, Laszczyńska M. Immunomodulatory Potential of Gut Microbiome-Derived Short-Chain Fatty Acids (SCFAs). Acta Biochim Pol (2019) 66(1):1–12. doi: 10.18388/ abp.2018_2648On behalf of the Obesity Programs of nutrition, Education, Research and Assessment (OPERA) group, Muscogiuri G, Cantone E, Cassarano S, Tuccinardi D, Barrea L, et al. Gut Microbiota: A New Path to Treat Obesity. Int J Obes Supp (2019) 9(1):10–9. doi: 10.1038/s41367-019-0011-7Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An Obesity-Associated Gut Microbiome With Increased Capacity for Energy Harvest. Nature (2006) 444(7122):1027–31. doi: 10.1038/nature05414Samuel BS, Shaito A, Motoike T, Rey FE, Backhed F, Manchester JK, et al. Effects of the Gut Microbiota on Host Adiposity Are Modulated by the Short-Chain Fatty-Acid Binding G Protein-Coupled Receptor, Gpr41. Proc Natl Acad Sci USA (2008) 105(43):16767–72. doi: 10.1073/pnas.0808567105Musso G, Gambino R, Cassader M. Interactions Between Gut Microbiota and Host Metabolism Predisposing to Obesity and Diabetes. Annu Rev Med (2011) 62:361–80. doi: 10.1146/annurev-med-012510-175505Grasset E, Puel A, Charpentier J, Collet X, Christensen JE, Tercé F, et al. A Specific Gut Microbiota Dysbiosis of Type 2 Diabetic Mice Induces GLP-1 Resistance Through an Enteric NO-Dependent and Gut-Brain Axis Mechanism. Cell Metab (2017) 26(1):278. doi: 10.1016/j.cmet.2017.04.013Schugar RC, Shih DM, Warrier M, Helsley RN, Burrows A, Ferguson D, et al. The TMAO-Producing Enzyme Flavin-Containing Monooxygenase 3 Regulates Obesity and the Beiging of White Adipose Tissue. Cell Rep (2017) 19(12):2451–61. doi: 10.1016/j.celrep.2017.05.077Cavallari JF, Fullerton MD, Duggan BM, Foley KP, Denou E, Smith BK, et al. Muramyl Dipeptide-Based Postbiotics Mitigate Obesity-Induced Insulin Resistance via IRF4. Cell Metab (2017) 25(5):1063–74.e3. doi: 10.1016/ j.cmet.2017.03.021Virtue AT, McCright SJ, Wright JM, Jimenez MT, Mowel WK, Kotzin JJ, et al. The Gut Microbiota Regulates White Adipose Tissue Inflammation and Obesity via a Family of microRNAs. Sci Transl Med (2019) 11(496): eaav1892. doi: 10.1126/scitranslmed.aav1892Patterson E, Ryan PM, Cryan JF, Dinan TG, Ross RP, Fitzgerald GF, et al. Gut Microbiota, Obesity and Diabetes. Postgrad Med J (2016) 92(1087):286– 300. doi: 10.1136/postgradmedj-2015-133285Martınez-Sa ́ ́nchez N. There and Back Again: Leptin Actions in White Adipose Tissue. Int J Mol Sci (2020) 21(17):6039. doi: 10.3390/ijms21176039Izquierdo AG, Crujeiras AB, Casanueva FF, Carreira MC. Leptin, Obesity, and Leptin Resistance: Where Are We 25 Years Later? Nutrients (2019) 11 (11):2704. doi: 10.3390/nu11112704Fuster JJ, Ouchi N, Gokce N, Walsh K. Obesity-Induced Changes in Adipose Tissue Microenvironment and Their Impact on Cardiovascular Disease. Circ Res (2016) 118(11):1786–807. doi: 10.1161/CIRCRESAHA.115.306885Teixeira TM, da Costa DC, Resende AC, Soulage CO, Bezerra FF, Daleprane JB. Activation of Nrf2-Antioxidant Signaling by 1,25- Dihydroxycholecalciferol Prevents Leptin-Induced Oxidative Stress and Inflammation in Human Endothelial Cells. J Nutr (2017) 147(4):506–13. doi: 10.3945/jn.116.239475Yamagishi SI, Edelstein D, Du XL, Kaneda Y, Guzmán M, Brownlee M. Leptin Induces Mitochondrial Superoxide Production and Monocyte Chemoattractant Protein-1 Expression in Aortic Endothelial Cells by Increasing Fatty Acid Oxidation via Protein Kinase a. J Biol Chem (2001) 276(27):25096–100. doi: 10.1074/jbc.M007383200Hongo S, Watanabe T, Arita S, Kanome T, Kageyama H, Shioda S, et al. Leptin Modulates ACAT1 Expression and Cholesterol Efflux From Human Macrophages. Am J Physiol Endocrinol Metab (2009) 297(2):E474–82. doi: 10.1152/ajpendo.90369.2008Singh S, Lohakare AC. Association of Leptin and Carotid Intima-Media Thickness in Overweight and Obese Individuals: A Cross-Sectional Study. J Assoc Physicians India (2020) 68(8):19–23. doi: 10.1155/2018/4285038Rahmani A, Toloueitabar Y, Mohsenzadeh Y, Hemmati R, Sayehmiri K, Asadollahi K. Association Between Plasma Leptin/Adiponectin Ratios With the Extent and Severity of Coronary Artery Disease. BMC Cardiovasc Disord (2020) 20(1):474. doi: 10.1186/s12872-020-01723-7Gan XT, Zhao G, Huang CX, Rowe AC, Purdham DM, Karmazyn M. Identification of Fat Mass and Obesity Associated (FTO) Protein Expression in Cardiomyocytes: Regulation by Leptin and Its Contribution to LeptinInduced Hypertrophy. PloS One (2013) 8(9):e74235. doi: 10.1371/ journal.pone.0074235Xu F-P, Chen M-S, Wang Y-Z, Yi Q, Lin S-B, Chen AF, et al. Leptin Induces Hypertrophy via Endothelin-1-Reactive Oxygen Species Pathway in Cultured Neonatal Rat Cardiomyocytes. Circulation (2004) 110(10):1269– 75. doi: 10.1161/01.CIR.0000140766.52771.6DZeidan A, Hunter JC, Javadov S, Karmazyn M. mTOR Mediates RhoADependent Leptin-Induced Cardiomyocyte Hypertrophy. Mol Cell Biochem (2011) 352(1):99–108. doi: 10.1007/s11010-011-0744-2Hou N, Luo M-S, Liu S-M, Zhang H-N, Xiao Q, Sun P, et al. Leptin Induces Hypertrophy Through Activating the Peroxisome Proliferator-Activated Receptor a Pathway in Cultured Neonatal Rat Cardiomyocytes. Clin Exp Pharmacol Physiol (2010) 37(11):1087–95. doi: 10.1111/j.1440-1681.2010. 05442.xPerego L, Pizzocri P, Corradi D, Maisano F, Paganelli M, Fiorina P, et al. Circulating Leptin Correlates With Left Ventricular Mass in Morbid (Grade III) Obesity Before and After Weight Loss Induced by Bariatric Surgery: A Potential Role for Leptin in Mediating Human Left Ventricular Hypertrophy. J Clin Endocrinol Metab (2005) 90(7):4087–93. doi: 10.1210/ jc.2004-1963Barouch Lili A, Berkowitz Dan E, Harrison Robert W, O’Donnell Christopher P, Hare Joshua M. Disruption of Leptin Signaling Contributes to Cardiac Hypertrophy Independently of Body Weight in Mice. Circulation (2003) 108(6):754–9. doi: 10.1161/01.CIR.0000083716.82622.FDMartin SS, Blaha MJ, Muse ED, Qasim AN, Reilly MP, Blumenthal RS, et al. Leptin and Incident Cardiovascular Disease: The Multi-Ethnic Study of Atherosclerosis (MESA). Atherosclerosis (2015) 239(1):67–72. doi: 10.1016/ j.atherosclerosis.2014.12.033Hall ME, Maready MW, Hall JE, Stec DE. Rescue of Cardiac Leptin Receptors in Db/Db Mice Prevents Myocardial Triglyceride Accumulation. Am J Physiol Endocrinol Metab (2014) 307(3):E316–25. doi: 10.1152/ ajpendo.00005.2014Lieb W, Xanthakis V, Sullivan LM, Aragam J, Pencina MJ, Larson MG, et al. Longitudinal Tracking of Left Ventricular Mass Over the Adult Life Course: Clinical Correlates of Short- and Long-Term Change in the Framingham Offspring Study. Circulation (2009) 119(24):3085–92. doi: 10.1161/ CIRCULATIONAHA.108.824243Mohamed-Ali V, Goodrick S, Rawesh A, Katz DR, Miles JM, Yudkin JS, et al. Subcutaneous Adipose Tissue Releases Interleukin-6, But Not Tumor Necrosis Factor-Alpha, In Vivo. J Clin Endocrinol Metab (1997) 82 (12):4196–200. doi: 10.1210/jcem.82.12.4450Yamauchi-Takihara K, Kishimoto T. Cytokines and Their Receptors in Cardiovascular Diseases–Role of Gp130 Signaling Pathway in Cardiac Myocyte Growth and Maintenance. Int J Exp Pathol (2000) 81(1):1–16. doi: 10.1046/j.1365-2613.2000.00139.xYu X-W, Chen Q, Kennedy RH, Liu SJ. Inhibition of Sarcoplasmic Reticular Function by Chronic Interleukin-6 Exposure via iNOS in Adult Ventricular Myocytes. J Physiol (2005) 566(Pt 2):327–40. doi: 10.1113/jphysiol.2005.086686Fontes JA, Rose NR, Č iháková D. The Varying Faces of IL-6: From Cardiac Protection to Cardiac Failure. Cytokine (2015) 74(1):62–8. doi: 10.1016/ j.cyto.2014.12.024Fuchs M, Hilfiker A, Kaminski K, Hilfiker-Kleiner D, Guener Z, Klein G, et al. Role of Interleukin-6 for LV Remodeling and Survival After Experimental Myocardial Infarction. FASEB J (2003) 17(14):2118–20. doi: 10.1096/fj.03-0331fje. Yan AT, Yan RT, Cushman M, Redheuil A, Tracy RP, Arnett DK, et al. Relationship of Interleukin-6 With Regional and Global Left-Ventricular Function in Asymptomatic Individuals Without Clinical Cardiovascular Disease: Insights From the Multi-Ethnic Study of Atherosclerosis. Eur Heart J (2010) 31(7):875–82. doi: 10.1093/eurheartj/ehp454Markousis-Mavrogenis G, Tromp J, Ouwerkerk W, Devalaraja M, Anker SD, Cleland JG, et al. The Clinical Significance of Interleukin-6 in Heart Failure: Results From the BIOSTAT-CHF Study. Eur J Heart Failure (2019) 21 (8):965–73. doi: 10.1002/ejhf.1482Hui X, Lam KSL, Vanhoutte PM, Xu A. Adiponectin and Cardiovascular Health: An Update. Br J Pharmacol (2012) 165(3):574–90. doi: 10.1111/ j.1476-5381.2011.01395.xDong F, Ren J. Adiponectin Improves Cardiomyocyte Contractile Function in Db/Db Diabetic Obese Mice. Obes (Silver Spring) (2009) 17(2):262–8. doi: 10.1038/oby.2008.545Amin RH, Mathews ST, Alli A, Leff T. Endogenously Produced Adiponectin Protects Cardiomyocytes From Hypertrophy by a Pparg-Dependent Autocrine Mechanism. Am J Physiol-Heart Circ Physiol (2010) 299(3): H690–8. doi: 10.1152/ajpheart.01032.2009Tao L, Gao E, Jiao X, Yuan Y, Li S, Christopher Theodore A, et al. Adiponectin Cardioprotection After Myocardial Ischemia/Reperfusion Involves the Reduction of Oxidative/Nitrative Stress. Circulation (2007) 115(11):1408–16. doi: 10.1161/CIRCULATIONAHA.106.666941Ikeda Y, Ohashi K, Shibata R, Pimentel DR, Kihara S, Ouchi N, et al. Cyclooxygenase-2 Induction by Adiponectin Is Regulated by a Sphingosine Kinase-1 Dependent Mechanism in Cardiac Myocytes. FEBS Lett (2008) 582 (7):1147–50. doi: 10.1016/j.febslet.2008.03.002Cao Y, Tao L, Yuan Y, Jiao X, Lau WB, Wang Y, et al. Endothelial Dysfunction in Adiponectin Deficiency and Its Mechanisms Involved. J Mol Cell Cardiol (2009) 46(3):413–9. doi: 10.1016/j.yjmcc.2008.10.014Pischon T. Plasma Adiponectin Levels and Risk of Myocardial Infarction in Men. JAMA (2004) 291(14):1730. doi: 10.1001/jama.291.14.1730Kozakova M, Muscelli E, Flyvbjerg A, Frystyk J, Morizzo C, Palombo C, et al. Adiponectin and Left Ventricular Structure and Function in Healthy Adults. J Clin Endocrinol Metab (2008) 93(7):2811–8. doi: 10.1210/jc.2007-2580Sook Lee E, Park S, Kim E, Sook Yoon Y, Ahn H-Y, Park C-Y, et al. Association Between Adiponectin Levels and Coronary Heart Disease and Mortality: A Systematic Review and Meta-Analysis. Int J Epidemiol (2013) 42 (4):1029–39. doi: 10.1093/ije/dyt087. Van Berendoncks AM, Garnier A, Beckers P, Hoymans VY, Possemiers N, Fortin D, et al. Functional Adiponectin Resistance at the Level of the Skeletal Muscle in Mild to Moderate Chronic Heart Failure. Circ Heart Fail (2010) 3 (2):185–94. doi: 10.1161/CIRCHEARTFAILURE.109.885525Menon V, Li L, Wang X, Greene T, Balakrishnan V, Madero M, et al. Adiponectin and Mortality in Patients With Chronic Kidney Disease. J Am Soc Nephrol (2006) 17(9):2599–606. doi: 10.1681/ASN.2006040331Sun H, Wang Y. Branched Chain Amino Acid Metabolic Reprogramming in Heart Failure. Biochim Biophys Acta (BBA) Mol Basis Dis (2016) 1862 (12):2270–5. doi: 10.1016/j.bbadis.2016.09.009Wang W, Zhang F, Xia Y, Zhao S, Yan W, Wang H, et al. Defective Branched Chain Amino Acid Catabolism Contributes to Cardiac Dysfunction and Remodeling Following Myocardial Infarction. Am J Physiol-Heart Circ Physiol (2016) 311(5):H1160–9. doi: 10.1152/ajpheart.00114.2016. Li T, Zhang Z, Kolwicz SC, Abell L, Roe ND, Kim M, et al. Defective Branched-Chain Amino Acid Catabolism Disrupts Glucose Metabolism and Sensitises the Heart to Ischemia-Reperfusion Injury. Cell Metab (2017) 25 (2):374–85. doi: 10.1016/j.cmet.2016.11.005Pisarenko OI. Mechanisms of Myocardial Protection by Amino Acids: Facts and Hypotheses. Clin Exp Pharmacol Physiol (1996) 23(8):627–33. doi: 10.1111/j.1440-1681.1996.tb01748.xHalama A, Horsch M, Kastenmüller G, Möller G, Kumar P, Prehn C, et al. Metabolic Switch During Adipogenesis: From Branched Chain Amino Acid Catabolism to Lipid Synthesis. Arch Biochem Biophysics (2016) 589:93–107. doi: 10.1016/j.abb.2015.09.013Herman MA, She P, Peroni OD, Lynch CJ, Kahn BB. Adipose Tissue Branched Chain Amino Acid (BCAA) Metabolism Modulates Circulating BCAA Levels*. J Biol Chem (2010) 285(15):11348–56. doi: 10.1074/ jbc.M109.075184Huang Y, Zhou M, Sun H, Wang Y. Branched-Chain Amino Acid Metabolism in Heart Disease: An Epiphenomenon or a Real Culprit? Cardiovasc Res (2011) 90(2):220–3. doi: 10.1093/cvr/cvr070Tobias DK, Lawler PR, Harada PH, Demler OV, Ridker PM, Manson JE, et al. Circulating Branched-Chain Amino Acids and Incident Cardiovascular Disease in a Prospective Cohort of US Women. Circ Genom Precis Med (2018) 11(4):e002157. doi: 10.1161/CIRCGEN.118.002157Peterson MB, Mead RJ, Welty JD. Free Amino Acids in Congestive Heart Failure. J Mol Cell Cardiol (1973) 5(2):139–47. doi: 10.1016/0022-2828(73) 90047-3Kato T, Niizuma S, Inuzuka Y, Kawashima T, Okuda J, Tamaki Y, et al. Analysis of Metabolic Remodeling in Compensated Left Ventricular Hypertrophy and Heart Failure. Circ Heart Fail (2010) 3(3):420–30. doi: 10.1161/CIRCHEARTFAILURE.109.888479Aquilani R, La Rovere M, Corbellini D, Pasini E, Verri M, Barbieri A, et al. Plasma Amino Acid Abnormalities in Chronic Heart Failure. Mechanisms, Potential Risks and Targets in Human Myocardium Metabolism. Nutrients (2017) 9(11):1251. doi: 10.3390/nu9111251Green CR, Wallace M, Divakaruni AS, Phillips SA, Murphy AN, Ciaraldi TP, et al. Branched-Chain Amino Acid Catabolism Fuels Adipocyte Differentiation and Lipogenesis. Nat Chem Biol (2016) 12(1):15–21. doi: 10.1038/nchembio.1961Thoonen R, Hindle AG, Scherrer-Crosbie M. Brown Adipose Tissue: The Heat Is on the Heart. Am J Physiol-Heart Circ Physiol (2016) 310(11):H1592– 605. doi: 10.1152/ajpheart.00698.2015Gruden G, Landi A, Bruno G. Natriuretic Peptides, Heart, and Adipose Tissue: New Findings and Future Developments for Diabetes Research. Dia Care (2014) 37(11):2899–908. doi: 10.2337/dc14-0669Saito Y, Nakao K, Itoh H, Yamada T, Mukoyama M, Arai H, et al. Brain Natriuretic Peptide Is a Novel Cardiac Hormone. Biochem Biophys Res Commun (1989) 158(2):360–8. doi: 10.1016/S0006-291X(89)80056-7Planavila A, Fernández-Solà J, Villarroya F. Cardiokines as Modulators of Stress-Induced Cardiac Disorders. In: Advances in Protein Chemistry and Structural Biology. Elsevier (2017). Available at: https://linkinghub.elsevier. com/retrieve/pii/S1876162317300020 (Accessed cited 2021 Mar 18).Shimano M, Ouchi N, Walsh K. Cardiokines: Recent Progress in Elucidating the Cardiac Secretome. Circulation (2012) 126(21):e327–e32. doi: 10.1161/ CIRCGEN.118.002157Hosoda K, Nakao K, Mukoyama M, Saito Y, Jougasaki M, Shirakami G, et al. Expression of Brain Natriuretic Peptide Gene in Human Heart. Production in the Ventricle. Hypertension (1991) 17(6_pt_2):1152–5. doi: 10.1161/ 01.hyp.17.6.1152Jahng JWS, Song E, Sweeney G. Crosstalk Between the Heart and Peripheral Organs in Heart Failure. Exp Mol Med (2016) 48(3):e217–7. doi: 10.1038/ emm.2016.20Sarzani P, Dessì-Fulgheri P, Paci VM, Espinosa E, Rappelli A. Expression of Natriuretic Peptide Receptors in Human Adipose and Other Tissues. J Endocrinol Invest (1996) 19(9):581–5. doi: 10.1007/BF03349021Bordicchia M, Liu D, Amri E-Z, Ailhaud G, Dessì-Fulgheri P, Zhang C, et al. Cardiac Natriuretic Peptides Act via P38 MAPK to Induce the Brown Fat Thermogenic Program in Mouse and Human Adipocytes. J Clin Invest (2012) 122(3):1022–36. doi: 10.1172/JCI59701Nakamura M, Sadoshima J. Heart Over Mind: Metabolic Control of White Adipose Tissue and Liver. EMBO Mol Med (2014) 6(12):1521–4. doi: 10.15252/emmm.201404749Dewey CM, Spitler KM, Ponce JM, Hall DD, Grueter CE. Cardiac-Secreted Factors as Peripheral Metabolic Regulators and Potential Disease Biomarkers. JAHA (2016) 5(6):e003101. doi: 10.1161/JAHA.115.003101Sengenes C, Stich V, Berlan M, Hejnova J, Lafontan M, Pariskova Z, et al. Increased Lipolysis in Adipose Tissue and Lipid Mobilisation to Natriuretic Peptides During Low-Calorie Diet in Obese Women. Int J Obes (2002) 26 (1):24–32. doi: 10.1038/sj.ijo.0801845Wang TJ, Larson MG, Levy D, Benjamin EJ, Leip EP, Wilson PWF, et al. Impact of Obesity on Plasma Natriuretic Peptide Levels. Circulation (2004) 109(5):594–600. doi: 10.1161/01.CIR.0000112582.16683.EAPlante E, Menaouar A, Danalache BA, Broderick TL, Jankowski M, Gutkowska J. Treatment With Brain Natriuretic Peptide Prevents the Development of Cardiac Dysfunction in Obese Diabetic Db/Db Mice. Diabetologia (2014) 57(6):1257–67. doi: 10.1007/s00125-014-3201-4Silva M. The Role of BNP on Adipose Tissue Adaptations. Eur J Heart Fail (2017) 19:5–601. doi: 10.1002/ejhf.833Kovacova Z, Tharp WG, Liu D, Wei W, Xie H, Collins S, et al. Adipose Tissue Natriuretic Peptide Receptor Expression Is Related to Insulin Sensitivity in Obesity and Diabetes: Natriuretic Peptide Receptors in Metabolic Disease. Obesity (2016) 24(4):820–8. doi: 10.1002/oby.21418Huang Y, Yan Y, Xv W, Qian G, Li C, Zou H, et al. A New Insight Into the Roles of MiRNAs in Metabolic Syndrome. BioMed Res Int (2018) 2018:1–15. doi: 10.1155/2018/7372636Aleksandrova K, Mozaffarian D, Pischon T. Addressing the Perfect Storm: Biomarkers in Obesity and Pathophysiology of Cardiometabolic Risk. Clin Chem (2018) 64(1):142–53. doi: 10.1373/clinchem.2017.275172Bang C, Antoniades C, Antonopoulos AS, Eriksson U, Franssen C, Hamdani N, et al. Intercellular Communication Lessons in Heart Failure: Communication in Heart Failure. Eur J Heart Fail (2015) 17(11):1091– 103. doi: 10.1002/ejhf.399Mazurek T, Opolski G. Pericoronary Adipose Tissue: A Novel Therapeutic Target in Obesity-Related Coronary Atherosclerosis. J Am Coll Nutr (2015) 34(3):244–54. doi: 10.1080/07315724.2014.933685Song Y, Song F, Wu C, Hong Y-X, Li G. The Roles of Epicardial Adipose Tissue in Heart Failure. Heart Fail Rev (2020). doi: 10.1007/s10741-020- 09997-x. Lambert C, Arderiu G, Bejar MT, Crespo J, Baldellou M, Juan-Babot O, et al. Stem Cells From Human Cardiac Adipose Tissue Depots Show Different Gene Expression and Functional Capacities. Stem Cell Res Ther (2019) 10 (1):361. doi: 10.1186/s13287-019-1460-1Vienberg S, Geiger J, Madsen S, Dalgaard LT. MicroRNAs in Metabolism. Acta Physiol (2017) 219(2):346–61. doi: 10.1111/apha.12681. Huang-Doran I, Zhang C-Y, Vidal-Puig A. Extracellular Vesicles: Novel Mediators of Cell Communication In Metabolic Disease. Trends Endocrinol Metab (2017) 28(1):3–18. doi: 10.1016/j.tem.2016.10.003. Zhang Y-F, Xu H-M, Yu F, Wang M, Li M-Y, Xu T, et al. Crosstalk Between MicroRNAs and Peroxisome Proliferator-Activated Receptors and Their Emerging Regulatory Roles in Cardiovascular Pathophysiology. PPAR Res (2018) 2018:1–11. doi: 10.1155/2018/8530371Ocłoń E, Latacz A, Zubel–Łojek J, Pierzchała–Koziec K. HyperglycemiaInduced Changes in miRNA Expression Patterns in Epicardial Adipose Tissue of Piglets. J Endocrinol (2016) 229(3):259–66. doi: 10.1530/JOE-15- 0495. Liu Y, Fu W, Lu M, Huai S, Song Y, Wei Y. Role of miRNAs in Epicardial Adipose Tissue in CAD Patients With T2DM. BioMed Res Int (2016) 2016:1–7. doi: 10.1155/2016/2816056Pan J, Alimujiang M, Chen Q, Shi H, Luo X. Exosomes Derived From miR146a-Modified Adipose-Derived Stem Cells Attenuate Acute Myocardial Infarction–Induced Myocardial Damage via Downregulation of Early Growth Response Factor 1. J Cell Biochem (2019) 120(3):4433–43. doi: 10.1002/jcb.27731Luo Q, Guo D, Liu G, Chen G, Hang M, Jin M. Exosomes From MiR-126- Overexpressing Adscs Are Therapeutic in Relieving Acute Myocardial Ischaemic Injury. Cell Physiol Biochem (2017) 44(6):2105–16. doi: 10.1159/ 000485949Zhai Y, Yang J, Zhang J, Yang J, Li Q, Zheng T. miRNA-3614 Derived From Epicardial Adipose Tissue: A Novel Target for Ischemic Heart Diseases. Int J Cardiol (2021) 326:157. doi: 10.1016/j.ijcard.2020.10.076Zou T, Zhu M, Ma Y-C, Xiao F, Yu X, Xu L, et al. MicroRNA-410-5p Exacerbates High-Fat Diet-Induced Cardiac Remodeling in Mice in an Endocrine Fashion. Sci Rep (2018) 8(1):8780. doi: 10.1038/s41598-018- 26646-4Bork-Jensen J, Thuesen A, Bang-Bertelsen C, Grunnet L, Pociot F, BeckNielsen H, et al. Genetic Versus Non-Genetic Regulation of miR-103, miR143 and miR-483-3p Expression in Adipose Tissue and Their Metabolic Implications—A Twin Study. Genes (2014) 5(3):508–17. doi: 10.3390/ genes5030508Scheja L, Heeren J. The Endocrine Function of Adipose Tissues in Health and Cardiometabolic Disease. Nat Rev Endocrinol (2019) 15(9):507–24. doi: 10.1038/s41574-019-0230-6Akoumianakis I, Antoniades C. The Interplay Between Adipose Tissue and the Cardiovascular System: Is Fat Always Bad? Cardiovasc Res (2017) 113 (9):999–1008. doi: 10.1093/cvr/cvx111Mormile R. The Obesity Paradox in Heart Failure: A miR-26b Affair? Int J Colorectal Dis (2017) 32(4):595–6. doi: 10.1007/s00384-017-2762-3Marı-Alexandre J, Barcelo ́ ́-Molina M, Sanz-Sánchez J, Molina P, Sancho J, Abellán Y, et al. Thickness and an Altered miRNA Expression in the Epicardial Adipose Tissue Is Associated With Coronary Heart Disease in Sudden Death Victims. Rev Española Cardiol (English Ed) (2019) 72(1):30–9. doi: 10.1016/j.rec.2017.12.007Araujo HN, Victório JA, Valgas da Silva CP, Sponton ACS, Vettorazzi JF, de Moraes C, et al. Anti-Contractile Effects of Perivascular Adipose Tissue in Thoracic Aorta From Rats Fed a High-Fat Diet: Role of Aerobic Exercise Training. Clin Exp Pharmacol Physiol (2018) 45(3):293–302. doi: 10.1111/ 1440-1681.12882Kang S, Kim KB, Shin KO. Exercise Training Improves Leptin Sensitivity in Peripheral Tissue of Obese Rats. Biochem Biophys Res Commun (2013) 435 (3):454–9. doi: 10.1016/j.bbrc.2013.05.007Su M, Bai Y-P, Song W-W, Wang M, Shen R-R, Du K-J, et al. Effect of Exercise on Adiponectin in Aged Obese Rats. Zhongguo Ying Yong Sheng Li Xue Za Zhi (2018) 34(4):345–9. doi: 10.12047/j.cjap.5660.2018.079Kazemi F, Zahediasl S. Effects of Exercise Training on Adipose Tissue Apelin Expression in Streptozotocin-Nicotinamide Induced Diabetic Rats. Gene (2018) 662:97–102. doi: 10.1016/j.gene.2018.04.003Meziat C, Boulghobra D, Strock E, Battault S, Bornard I, Walther G, et al. Exercise Training Restores eNOS Activation in the Perivascular Adipose Tissue of Obese Rats: Impact on Vascular Function. Nitric Oxide (2019) 86:63–7. doi: 10.1016/j.niox.2019.02.009Khoo J, Dhamodaran S, Chen D-D, Yap S-Y, Chen RY-T, Tian RH-H. Exercise-Induced Weight Loss Is More Effective Than Dieting for Improving Adipokine Profile, Insulin Resistance, and Inflammation in Obese Men. Int J Sport Nutr Exerc Metab (2015) 25(6):566–75. doi: 10.1123/ijsnem.2015-0025Abd El-Kader SM, Al-Jiffri OH, Neamatallah ZA, AlKhateeb AM, AlFawaz SS. Weight Reduction Ameliorates Inflammatory Cytokines, Adipocytokines and Endothelial Dysfunction Biomarkers Among Saudi Patients With Type 2 Diabetes. Afr Health Sci (2020) 20(3):1329–36. doi: 10.4314/ahs.v20i3.39Elloumi M, Ben Ounis O, Makni E, Van Praagh E, Tabka Z, Lac G. Effect of Individualised Weight-Loss Programmes on Adiponectin, Leptin and Resistin Levels in Obese Adolescent Boys. Acta Paediatr (2009) 98 (9):1487–93. doi: 10.1111/j.1651-2227.2009.01365.xMaillard F, Pereira B, Boisseau N. Effect of High-Intensity Interval Training on Total, Abdominal and Visceral Fat Mass: A Meta-Analysis. Sports Med (2018) 48(2):269–88. doi: 10.1007/s40279-017-0807-yWewege M, van den Berg R, Ward RE, Keech A. The Effects of HighIntensity Interval Training vs. Moderate-Intensity Continuous Training on Body Composition in Overweight and Obese Adults: A Systematic Review and Meta-Analysis. Obes Rev (2017) 18(6):635–46. doi: 10.1111/obr.12532Párrizas M, Brugnara L, Esteban Y, González-Franquesa A, Canivell S, Murillo S, et al. Circulating miR-192 and miR-193b Are Markers of Prediabetes and Are Modulated by an Exercise Intervention. J Clin Endocrinol Metab (2015) 100(3):E407–415. doi: 10.1210/jc.2014-2574Russo A, Bartolini D, Mensà E, Torquato P, Albertini MC, Olivieri F, et al. Physical Activity Modulates the Overexpression of the Inflammatory miR146a-5p in Obese Patients. IUBMB Life (2018) 70(10):1012–22. doi: 10.1002/ iub.1926Amor S, Martın-Carro B, Rubio C, Carrascosa JM, Hu W, Huang Y, et al. ́ Study of Insulin Vascular Sensitivity in Aortic Rings and Endothelial Cells From Aged Rats Subjected to Caloric Restriction: Role of Perivascular Adipose Tissue. Exp Gerontol (2018) 109:126–36. doi: 10.1016/j.exger. 2017.10.017Bussey CE, Withers SB, Aldous RG, Edwards G, Heagerty AM. ObesityRelated Perivascular Adipose Tissue Damage Is Reversed by Sustained Weight Loss in the Rat. Arterioscler Thromb Vasc Biol (2016) 36(7):1377– 85. doi: 10.1161/ATVBAHA.116.307210Kim K-H, Kim YH, Son JE, Lee JH, Kim S, Choe MS, et al. Intermittent Fasting Promotes Adipose Thermogenesis and Metabolic Homeostasis via VEGF-Mediated Alternative Activation of Macrophage. Cell Res (2017) 27 (11):1309–26. doi: 10.1038/cr.2017.126Klempel MC, Kroeger CM, Bhutani S, Trepanowski JF, Varady KA. Intermittent Fasting Combined With Calorie Restriction Is Effective for Weight Loss and Cardio-Protection in Obese Women. Nutr J (2012) 11:98. doi: 10.1186/1475-2891-11-98Kroeger CM, Klempel MC, Bhutani S, Trepanowski JF, Tangney CC, Varady KA. Improvement in Coronary Heart Disease Risk Factors During an Intermittent Fasting/Calorie Restriction Regimen: Relationship to Adipokine Modulations. Nutr Metab (Lond) (2012) 9(1):98. doi: 10.1186/ 1743-7075-9-98Trepanowski JF, Kroeger CM, Barnosky A, Klempel M, Bhutani S, Hoddy KK, et al. Effects of Alternate-Day Fasting or Daily Calorie Restriction on Body Composition, Fat Distribution, and Circulating Adipokines: Secondary Analysis of a Randomised Controlled Trial. Clin Nutr (2018) 37(6 Pt A):1871–8. doi: 10.1016/j.clnu.2017.11.018Hsieh C-H, Rau C-S, Wu S-C, Yang JC-S, Wu Y-C, Lu T-H, et al. WeightReduction Through a Low-Fat Diet Causes Differential Expression of Circulating microRNAs in Obese C57BL/6 Mice. BMC Genomics (2015) 16(1):699. doi: 10.1186/s12864-015-1896-3Manning P, Munasinghe PE, Bellae Papannarao J, Gray AR, Sutherland W, Katare R. Acute Weight Loss Restores Dysregulated Circulating MicroRNAs in Individuals Who Are Obese. J Clin Endocrinol Metab (2019) 104(4):1239– 48. doi: 10.1210/jc.2018-00684Tabet F, Cuesta Torres LF, Ong KL, Shrestha S, Choteau SA, Barter PJ, et al. High-Density Lipoprotein-Associated miR-223 Is Altered After DietInduced Weight Loss in Overweight and Obese Males. PloS One (2016) 11 (3):e0151061. doi: 10.1371/journal.pone.0151061Bagarolli RA, Tobar N, Oliveira AG, Araújo TG, Carvalho BM, Rocha GZ, et al. Probiotics Modulate Gut Microbiota and Improve Insulin Sensitivity in DIO Mice. J Nutr Biochem (2017) 50:16–25. doi: 10.1016/j.jnutbio. 2017.08.006Behrouz V, Jazayeri S, Aryaeian N, Zahedi MJ, Hosseini F. Effects of Probiotic and Prebiotic Supplementation on Leptin, Adiponectin, and Glycemic Parameters in Non-Alcoholic Fatty Liver Disease: A Randomised Clinical Trial. Middle East J Dig Dis (2017) 9(3):150–7. doi: 10.15171/ mejdd.2017.66Al-Muzafar HM, Amin KA. Probiotic Mixture Improves Fatty Liver Disease by Virtue of Its Action on Lipid Profiles, Leptin, and Inflammatory Biomarkers. BMC Complement Altern Med (2017) 17(1):43. doi: 10.1186/ s12906-016-1540-zBernini LJ, Simão ANC, de Souza CHB, Alfieri DF, Segura LG, Costa GN, et al. Effect of Bifidobacterium Lactis HN019 on Inflammatory Markers and Oxidative Stress in Subjects With and Without the Metabolic Syndrome. Br J Nutr (2018) 120(6):645–52. doi: 10.1017/S0007114518001861Sabico S, Al-Mashharawi A, Al-Daghri NM, Wani K, Amer OE, Hussain DS, et al. Effects of a 6-Month Multi-Strain Probiotics Supplementation in Endotoxemic, Inflammatory and Cardiometabolic Status of T2DM Patients: A Randomised, Double-Blind, Placebo-Controlled Trial. Clin Nutr (2019) 38(4):1561–9. doi: 10.1016/j.clnu.2018.08.009Karimi E, Moini A, Yaseri M, Shirzad N, Sepidarkish M, Hossein-Boroujerdi M, et al. Effects of Synbiotic Supplementation on Metabolic Parameters and Apelin in Women With Polycystic Ovary Syndrome: A Randomised DoubleBlind Placebo-Controlled Trial. Br J Nutr (2018) 119(4):398–406. doi: 10.1017/S0007114517003920Gan XT, Ettinger G, Huang CX, Burton JP, Haist JV, Rajapurohitam V, et al. Probiotic Administration Attenuates Myocardial Hypertrophy and Heart Failure After Myocardial Infarction in the Rat. Circ Heart Fail (2014) 7 (3):491–9. doi: 10.1161/CIRCHEARTFAILURE.113.000978. Huang F, Del-Rıo-Navarro BE, Leija-Martinez J, Torres-Alcantara S, Ruiz- ́ Bedolla E, Hernández-Cadena L, et al. Effect of Omega-3 Fatty Acids Supplementation Combined With Lifestyle Intervention on Adipokines and Biomarkers of Endothelial Dysfunction in Obese Adolescents With Hypertriglyceridemia. J Nutr Biochem (2019) 64:162–9. doi: 10.1016/ j.jnutbio.2018.10.012Mostowik M, Gajos G, Zalewski J, Nessler J, Undas A. Omega-3 Polyunsaturated Fatty Acids Increase Plasma Adiponectin to Leptin Ratio in Stable Coronary Artery Disease. Cardiovasc Drugs Ther (2013) 27(4):289– 95. doi: 10.1007/s10557-013-6457-xJafari Salim S, Alizadeh S, Djalali M, Nematipour E, Hassan Javanbakht M. Effect of Omega-3 Polyunsaturated Fatty Acids Supplementation on Body Composition and Circulating Levels of Follistatin-Like 1 in Males With Coronary Artery Disease: A Randomised Double-Blind Clinical Trial. Am J Mens Health (2017) 11(6):1758–64. doi: 10.1177/1557988317720581Wang C, Xiong B, Huang J. The Role of Omega-3 Polyunsaturated Fatty Acids in Heart Failure: A Meta-Analysis of Randomised Controlled Trials. Nutrients (2016) 9(1):18. doi: 10.3390/nu9010018Redondo Useros N, Gheorghe A, Perez de Heredia F, Dıaz LE, Baccan GC, de ́ la Fuente M, et al. 2-OHOA Supplementation Reduced Adiposity and Improved Cardiometabolic Risk to a Greater Extent Than N-3 PUFA in Obese Mice. Obes Res Clin Pract (2019) 13(6):579–85. doi: 10.1016/ j.orcp.2019.10.009Luvizotto R de AM, Nascimento AF, Imaizumi E, Pierine DT, Conde SJ, Correa CR, et al. Lycopene Supplementation Modulates Plasma Concentrations and Epididymal Adipose Tissue mRNA of Leptin, Resistin and IL-6 in Diet-Induced Obese Rats. Br J Nutr (2013) 110(10):1803–9. doi: 10.1017/S0007114513001256Clark CCT, Ghaedi E, Arab A, Pourmasoumi M, Hadi A. The Effect of Curcumin Supplementation on Circulating Adiponectin: A Systematic Review and Meta-Analysis of Randomised Controlled Trials. Diabetes Metab Syndr (2019) 13(5):2819–25. doi: 10.1016/j.dsx.2019.07.045. Shamsi-Goushki A, Mortazavi Z, Mirshekar MA, Mohammadi M, MoradiKor N, Jafari-Maskouni S, et al. Comparative Effects of Curcumin Versus Nano-Curcumin on Insulin Resistance, Serum Levels of Apelin and Lipid Profile in Type 2 Diabetic Rats. Diabetes Metab Syndr Obes (2020) 13:2337– 46. doi: 10.2147/DMSO.S247351Mohammadi-Sartang M, Mazloom Z, Sohrabi Z, Sherafatmanesh S, BaratiBoldaji R. Resveratrol Supplementation and Plasma Adipokines Concentrations? A Systematic Review and Meta-Analysis of Randomised Controlled Trials. Pharmacol Res (2017) 117:394–405. doi: 10.1016/ j.phrs.2017.01.012Pérez de Heredia F, Wood IS, Trayhurn P. Hypoxia Stimulates Lactate Release and Modulates Monocarboxylate Transporter (MCT1, MCT2, and MCT4) Expression in Human Adipocytes. Pflugers Arch (2010) 459(3):509– 18. doi: 10.1007/s00424-009-0750-3CC-LICENSElicense_rdflicense_rdfapplication/rdf+xml; charset=utf-8805https://bonga.unisimon.edu.co/bitstreams/0f4257c3-18db-438b-b158-5f9c98298d90/download4460e5956bc1d1639be9ae6146a50347MD52LICENSElicense.txtlicense.txttext/plain; charset=utf-8381https://bonga.unisimon.edu.co/bitstreams/b5967e31-bd03-43cf-bdc7-b2789bf55486/download733bec43a0bf5ade4d97db708e29b185MD53ORIGINALPDF.pdfPDF.pdfPDFapplication/pdf942005https://bonga.unisimon.edu.co/bitstreams/3c031e5e-d23b-43b8-8862-b15475786801/download99ded17c699dd710b771919833abd341MD51TEXT2021_FinE_The-Sick-Adipose-Tissue.pdf.txt2021_FinE_The-Sick-Adipose-Tissue.pdf.txtExtracted texttext/plain102954https://bonga.unisimon.edu.co/bitstreams/b1389a5a-1e9b-4f56-ba06-85a7c6fa1eee/download763b57de281c64f954cdc0b386be58ccMD54PDF.pdf.txtPDF.pdf.txtExtracted texttext/plain100475https://bonga.unisimon.edu.co/bitstreams/98055058-2c13-4ac6-ad81-5bfd7d83a222/downloadd497161139e30a3de3dd9e26e116d5fcMD56THUMBNAIL2021_FinE_The-Sick-Adipose-Tissue.pdf.jpg2021_FinE_The-Sick-Adipose-Tissue.pdf.jpgGenerated Thumbnailimage/jpeg18762https://bonga.unisimon.edu.co/bitstreams/7dd81a2e-8d31-4335-9f56-e1ada6eb6518/downloadee248de9193d6c622d195731ebadd122MD55PDF.pdf.jpgPDF.pdf.jpgGenerated Thumbnailimage/jpeg5601https://bonga.unisimon.edu.co/bitstreams/fa36acde-3b7f-46dc-818c-c47cf5a7d427/download1934ac2cd8b3eb6ec920d98989665f28MD5720.500.12442/8657oai:bonga.unisimon.edu.co:20.500.12442/86572024-08-14 21:54:49.435http://creativecommons.org/licenses/by-nc-nd/4.0/Attribution-NonCommercial-NoDerivatives 4.0 Internacionalopen.accesshttps://bonga.unisimon.edu.coRepositorio Digital Universidad Simón Bolívarrepositorio.digital@unisimon.edu.coPGEgcmVsPSJsaWNlbnNlIiBocmVmPSJodHRwOi8vY3JlYXRpdmVjb21tb25zLm9yZy9saWNlbnNlcy9ieS1uYy80LjAvIj48aW1nIGFsdD0iTGljZW5jaWEgQ3JlYXRpdmUgQ29tbW9ucyIgc3R5bGU9ImJvcmRlci13aWR0aDowO3dpZHRoOjEwMHB4OyIgc3JjPSJodHRwczovL2kuY3JlYXRpdmVjb21tb25zLm9yZy9sL2J5LW5jLzQuMC84OHgzMS5wbmciIC8+PC9hPjxici8+RXN0YSBvYnJhIGVzdMOhIGJham8gdW5hIDxhIHJlbD0ibGljZW5zZSIgaHJlZj0iaHR0cDovL2NyZWF0aXZlY29tbW9ucy5vcmcvbGljZW5zZXMvYnktbmMvNC4wLyI+TGljZW5jaWEgQ3JlYXRpdmUgQ29tbW9ucyBBdHJpYnVjacOzbi1Ob0NvbWVyY2lhbCA0LjAgSW50ZXJuYWNpb25hbDwvYT4u