1
|
Zhang F, Zhou J, Wang L, Zeng Z, Fu S, Xie P, Meng X. Residual abnormalities on CTE predict adverse outcomes in Crohn's disease with endoscopic healing. Dig Liver Dis 2024; 56:248-257. [PMID: 37758612 DOI: 10.1016/j.dld.2023.09.008] [Citation(s) in RCA: 2] [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] [Received: 06/16/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Residual abnormalities on computed tomography enterography (CTE) in Crohn's disease (CD) with endoscopic healing (EH) may have prognostic implications and affect therapeutic strategy. METHODS CD patients with EH who underwent CTE between March 2015 and June 2022 were enrolled. CTE findings of the terminal ileum and the most severe segment of colon at the time of EH were assessed respectively for each patient. Cox regression analysis and Kaplan-Meier curves were used to evaluate the association between residual abnormalities and adverse outcomes. RESULTS A total of 140 patients (217 digestive segments) were included. Mesenteric edema (hazard ratio [HR] = 3.61, 95% CI = 1.81-7.20, P<0.001), fibrofatty proliferation (HR = 3.40, 95% CI = 1.97-5.85, P<0.001) and active small bowel inflammation (HR = 2.74, 95% CI = 1.59-4.71, P<0.001) were risk factors for clinical relapse. Furthermore, we built a scoring system using the three parameters. Radiologic score ≥ 1 was the best threshold to predict clinical relapse (HR = 4.56, 95% CI = 2.54-8.19, P<0.001) and it was validated in different outcomes. CONCLUSION The scoring system based on three residual abnormalities on CTE can predict adverse outcomes in CD patients with EH.
Collapse
Affiliation(s)
- Fangling Zhang
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University. 26th Yuancun the second Road, Guangzhou, Guangdong Province, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, China
| | - Jie Zhou
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University. 26th Yuancun the second Road, Guangzhou, Guangdong Province, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, China
| | - Ling Wang
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University. 26th Yuancun the second Road, Guangzhou, Guangdong Province, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, China
| | - Zhiming Zeng
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University. 26th Yuancun the second Road, Guangzhou, Guangdong Province, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, China
| | - Shuai Fu
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University. 26th Yuancun the second Road, Guangzhou, Guangdong Province, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, China
| | - Peiyi Xie
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University. 26th Yuancun the second Road, Guangzhou, Guangdong Province, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, China.
| | - Xiaochun Meng
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University. 26th Yuancun the second Road, Guangzhou, Guangdong Province, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, China.
| |
Collapse
|
2
|
Li X, Ma L. From biological aging to functional decline: Insights into chronic inflammation and intrinsic capacity. Ageing Res Rev 2024; 93:102175. [PMID: 38145874 DOI: 10.1016/j.arr.2023.102175] [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: 05/18/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023]
Abstract
Intrinsic capacity is the sum of an individual's physical and mental capacities, which helps determine functional ability. Intrinsic capacity decline is an important predictor of adverse health outcomes and can identify individuals at higher risk of functional decline. Aging is characterized by a decrease in physiological reserves and functional abilities. Chronic inflammation, a mechanism of aging, is associated with decreased intrinsic capacity, which may mirror the broader relationship between aging and functional ability. Therefore, it is crucial for maintaining functional ability and promoting healthy aging to study the mechanisms of intrinsic capacity decline, identify easily available markers, and make targets for intervention from the perspective of chronic inflammation. We reviewed the current research on chronic inflammation, inflammation-related markers, and intrinsic capacity. To date, there is still no inflammatory markers with high specificity and sensitivity to monitor intrinsic capacity decline. Interleukin-6, C-reactive protein, and tumor necrosis factor-alpha may potentially indicate changes in intrinsic capacity, but their results with intrinsic capacity or each intrinsic capacity domain are inconsistent. Considering the variations in individual responses to changes in inflammatory markers, it may be beneficial to explore the use of multiple analytes instead of relying on a single marker. This approach could be valuable in monitoring the decline of intrinsic capacity in the future.
Collapse
Affiliation(s)
- Xiaxia Li
- Department of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Lina Ma
- Department of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders, Beijing, China.
| |
Collapse
|
3
|
McCall KD, Walter D, Patton A, Thuma JR, Courreges MC, Palczewski G, Goetz DJ, Bergmeier S, Schwartz FL. Anti-Inflammatory and Therapeutic Effects of a Novel Small-Molecule Inhibitor of Inflammation in a Male C57BL/6J Mouse Model of Obesity-Induced NAFLD/MAFLD. J Inflamm Res 2023; 16:5339-5366. [PMID: 38026235 PMCID: PMC10658948 DOI: 10.2147/jir.s413565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Non-alcoholic fatty liver disease (NAFLD), recently renamed metabolic (dysfunction) associated fatty liver disease (MAFLD), is the most common chronic liver disease in the United States. Presently, there is an intense and ongoing effort to identify and develop novel therapeutics for this disease. In this study, we explored the anti-inflammatory activity of a new compound, termed IOI-214, and its therapeutic potential to ameliorate NAFLD/MAFLD in male C57BL/6J mice fed a high fat (HF) diet. Methods Murine macrophages and hepatocytes in culture were treated with lipopolysaccharide (LPS) ± IOI-214 or DMSO (vehicle), and RT-qPCR analyses of inflammatory cytokine gene expression were used to assess IOI-214's anti-inflammatory properties in vitro. Male C57BL/6J mice were also placed on a HF diet and treated once daily with IOI-214 or DMSO for 16 weeks. Tissues were collected and analyzed to determine the effects of IOI-214 on HF diet-induced NAFL D/MAFLD. Measurements such as weight, blood glucose, serum cholesterol, liver/serum triglyceride, insulin, and glucose tolerance tests, ELISAs, metabolomics, Western blots, histology, gut microbiome, and serum LPS binding protein analyses were conducted. Results IOI-214 inhibited LPS-induced inflammation in macrophages and hepatocytes in culture and abrogated HF diet-induced mesenteric fat accumulation, hepatic inflammation and steatosis/hepatocellular ballooning, as well as fasting hyperglycemia without affecting insulin resistance or fasting insulin, cholesterol or TG levels despite overall obesity in vivo in male C57BL/6J mice. IOI-214 also decreased systemic inflammation in vivo and improved gut microbiota dysbiosis and leaky gut. Conclusion Combined, these data indicate that IOI-214 works at multiple levels in parallel to inhibit the inflammation that drives HF diet-induced NAFLD/MAFLD, suggesting that it may have therapeutic potential for NAFLD/MAFLD.
Collapse
Affiliation(s)
- Kelly D McCall
- Molecular and Cellular Biology Program, Ohio University College of Arts & Sciences, Athens, OH, USA
- Department of Biological Sciences, Ohio University College of Arts & Sciences, Athens, OH, USA
- Department of Specialty Medicine, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Biomedical Engineering Program, Ohio University Russ College of Engineering and Technology, Athens, OH, USA
| | - Debra Walter
- Molecular and Cellular Biology Program, Ohio University College of Arts & Sciences, Athens, OH, USA
- Department of Biological Sciences, Ohio University College of Arts & Sciences, Athens, OH, USA
| | - Ashley Patton
- Molecular and Cellular Biology Program, Ohio University College of Arts & Sciences, Athens, OH, USA
- Department of Biological Sciences, Ohio University College of Arts & Sciences, Athens, OH, USA
| | - Jean R Thuma
- Department of Specialty Medicine, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
| | - Maria C Courreges
- Department of Specialty Medicine, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
| | | | - Douglas J Goetz
- Molecular and Cellular Biology Program, Ohio University College of Arts & Sciences, Athens, OH, USA
- Biomedical Engineering Program, Ohio University Russ College of Engineering and Technology, Athens, OH, USA
- Department of Chemical & Biomolecular Engineering, Ohio University Russ College of Engineering and Technology, Athens, OH, USA
| | - Stephen Bergmeier
- Molecular and Cellular Biology Program, Ohio University College of Arts & Sciences, Athens, OH, USA
- Biomedical Engineering Program, Ohio University Russ College of Engineering and Technology, Athens, OH, USA
- Department of Chemistry & Biochemistry, Ohio University College of Arts & Sciences, Athens, OH, USA
| | - Frank L Schwartz
- Department of Specialty Medicine, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Biomedical Engineering Program, Ohio University Russ College of Engineering and Technology, Athens, OH, USA
| |
Collapse
|
4
|
Tsounis EP, Aggeletopoulou I, Mouzaki A, Triantos C. Creeping Fat in the Pathogenesis of Crohn's Disease: An Orchestrator or a Silent Bystander? Inflamm Bowel Dis 2023; 29:1826-1836. [PMID: 37260352 DOI: 10.1093/ibd/izad095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Indexed: 06/02/2023]
Abstract
Although the phenomenon of hypertrophied adipose tissue surrounding inflamed bowel segments in Crohn's disease has been described since 1932, the mechanisms mediating the creeping fat formation and its role in the pathogenesis of the disease have not been fully unraveled. Recent advances demonstrating the multiple actions of adipose tissue beyond energy storage have brought creeping fat to the forefront of scientific research. In Crohn's disease, dysbiosis and transmural injury compromise the integrity of the intestinal barrier, resulting in an excessive influx of intraluminal microbiota and xenobiotics. The gut and peri-intestinal fat are in close anatomic relationship, implying a direct reciprocal immunologic relationship, whereas adipocytes are equipped with an arsenal of innate immunity sensors that respond to invading stimuli. As a result, adipocytes and their progenitor cells undergo profound immunophenotypic changes, leading to adipose tissue remodeling and eventual formation of creeping fat. Indeed, creeping fat is an immunologically active organ that synthesizes various pro- and anti-inflammatory cytokines, profibrotic mediators, and adipokines that serve as paracrine/autocrine signals and regulate immune responses. Therefore, creeping fat appears to be involved in inflammatory signaling, which explains why it has been associated with a higher severity or complicated phenotype of Crohn's disease. Interestingly, there is growing evidence for an alternative immunomodulatory function of creeping fat as a second barrier that prevents an abnormal systemic inflammatory response at the expense of an increasingly proliferating profibrotic environment. Further studies are needed to clarify how this modified adipose tissue exerts its antithetic effect during the course of Crohn's disease.
Collapse
Affiliation(s)
- Efthymios P Tsounis
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, Patras 26504, Greece
| | - Ioanna Aggeletopoulou
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, Patras 26504, Greece
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Athanasia Mouzaki
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, Patras 26504, Greece
| |
Collapse
|
5
|
Zhou Z, Zhang H, Tao Y, Zang J, Zhao J, Li H, Wang Y, Wang T, Zhao H, Wang F, Guo C, Zhu F, Mao H, Liu F, Zhang L, Wang Q. FGF21 alleviates adipose stem cell senescence via CD90 glycosylation-dependent glucose influx in remodeling healthy white adipose tissue. Redox Biol 2023; 67:102877. [PMID: 37690164 PMCID: PMC10497791 DOI: 10.1016/j.redox.2023.102877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/04/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023] Open
Abstract
The senescence of adipose stem cells (ASCs) impairs healthy adipose tissue remodeling, causing metabolic maladaptation to energy surplus. The intrinsic molecular pathways and potential therapy targets for ASC senescence are largely unclear. Here, we showed that visceral ASCs were prone to senescence that was caused by reactive oxygen species (ROS) overload, especially mitochondrial ROS. These senescent ASCs failed to sustain efficient glucose influx, pentose phosphate pathway (PPP) and redox homeostasis. We showed that CD90 silence restricted the glucose uptake by ASCs and thus disrupted their PPP and anti-oxidant system, resulting in ASC senescence. Notably, fibroblast growth factor 21 (FGF21) treatment significantly reduced the senescent phenotypes of ASCs by augmenting CD90 protein via glycosylation, which promoted glucose influx via the AKT-GLUT4 axis and therefore mitigated ROS overload. For diet-induced obese mice, chronic administration of low-dose FGF21 relieved their visceral white adipose tissue (VAT) dysfunction and systemic metabolic disorders. In particular, VAT homeostasis was restored in FGF21-treated obese mice, where ASC repertoire was markedly recovered, accompanied by CD90 elevation and anti-senescent phenotypes in these ASCs. Collectively, we reveal a molecular mechanism of ASC senescence by which CD90 downregulation interferes glucose influx into PPP and redox homeostasis. And we propose a FGF21-based strategy for healthy VAT remodeling, which targets CD90 glycosylation to correct ASC senescence and therefore combat obesity-related metabolic dysfunction.
Collapse
Affiliation(s)
- Zixin Zhou
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Huiying Zhang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yan Tao
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Jinhao Zang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Jingyuan Zhao
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Huijie Li
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yalin Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Tianci Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Hui Zhao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Fuwu Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chun Guo
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Faliang Zhu
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Haiting Mao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Fengming Liu
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Lining Zhang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
6
|
Al Madhoun A, Kochumon S, Haddad D, Thomas R, Nizam R, Miranda L, Sindhu S, Bitar MS, Ahmad R, Al-Mulla F. Adipose Tissue Caveolin-1 Upregulation in Obesity Involves TNF-α/NF-κB Mediated Signaling. Cells 2023; 12:cells12071019. [PMID: 37048092 PMCID: PMC10093236 DOI: 10.3390/cells12071019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 03/19/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Obesity is characterized by chronic low-grade inflammation. Obese people have higher levels of caveolin-1 (CAV1), a structural and functional protein present in adipose tissues (ATs). We aimed to define the inflammatory mediators that influence CAV1 gene regulation and the associated mechanisms in obesity. Using subcutaneous AT from 27 (7 lean and 20 obese) normoglycemic individuals, in vitro human adipocyte models, and in vivo mice models, we found elevated CAV1 expression in obese AT and a positive correlation between the gene expression of CAV1, tumor necrosis factor-alpha (TNF-α), and the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). CAV1 gene expression was associated with proinflammatory cytokines and chemokines and their cognate receptors (r ≥ 0.447, p ≤ 0.030), but not with anti-inflammatory markers. CAV1 expression was correlated with CD163, indicating a prospective role for CAV1 in the adipose inflammatory microenvironment. Unlike wild-type animals, mice lacking TNF-α exhibited reduced levels of CAV1 mRNA/proteins, which were elevated by administering exogenous TNF-α. Mechanistically, TNF-α induces CAV1 gene transcription by mediating NF-κB binding to its two regulatory elements located in the CAV1 proximal regulatory region. The interplay between CAV1 and the TNF-α signaling pathway is intriguing and has potential as a target for therapeutic interventions in obesity and metabolic syndromes.
Collapse
Affiliation(s)
- Ashraf Al Madhoun
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (R.N.); (M.S.B.)
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait; (L.M.); (S.S.)
- Correspondence: (A.A.M.); (R.A.); (F.A.-M.); Tel.: +965-2224-2999 (ext. 2211) (F.A.-M.)
| | - Shihab Kochumon
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (R.T.)
| | - Dania Haddad
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (R.N.); (M.S.B.)
| | - Reeby Thomas
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (R.T.)
| | - Rasheeba Nizam
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (R.N.); (M.S.B.)
| | - Lavina Miranda
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait; (L.M.); (S.S.)
| | - Sardar Sindhu
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait; (L.M.); (S.S.)
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (R.T.)
| | - Milad S. Bitar
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (R.N.); (M.S.B.)
- Department of Pharmacology, Faculty of Medicine, Kuwait University, Jabriya 046300, Kuwait
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (R.T.)
- Correspondence: (A.A.M.); (R.A.); (F.A.-M.); Tel.: +965-2224-2999 (ext. 2211) (F.A.-M.)
| | - Fahd Al-Mulla
- Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (R.N.); (M.S.B.)
- Correspondence: (A.A.M.); (R.A.); (F.A.-M.); Tel.: +965-2224-2999 (ext. 2211) (F.A.-M.)
| |
Collapse
|
7
|
Gomes SV, Dias BV, Júnior PAM, Pereira RR, de Souza DMS, Breguez GS, de Lima WG, Magalhães CLDB, Cangussú SD, Talvani A, Queiroz KB, Calsavara AJC, Costa DC. High-fat diet increases mortality and intensifies immunometabolic changes in septic mice. J Nutr Biochem 2023; 116:109315. [PMID: 36921735 DOI: 10.1016/j.jnutbio.2023.109315] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/21/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023]
Abstract
Immunometabolic changes in the liver and white adipose tissue (WAT) caused by high-fat (HF) diet intake may worse metabolic adaptation and protection against pathogens in sepsis. We investigate the effect of chronic HF diet (15 weeks) on mortality and immunometabolic responses in female mice after sepsis induced by cecum ligation and perforation (CLP). At week 14, animals were divided into four groups: sham C diet (C-Sh), sepsis C diet (C-Sp), sham HF diet (HF-Sh) and sepsis HF diet (HF-Sp). The surviving animals were euthanised on the 7th day. The HF diet decreased survival rate (58.3% vs 76.2% C-Sp group), increased serum cytokine storm (IL-6 (1.41 ×; vs HF-Sh), IL-1β (1.37 ×; vs C-Sp), TNF (1.34 ×; vs C-Sp and 1.72 ×; vs HF-Sh), IL-17 (1.44 ×; vs HF-Sh), IL-10 (1.55 ×; vs C-Sp and 1.41 ×; HF-Sh), WAT inflammation (IL-6 (8.7 ×; vs C-Sp and 2.4 ×; vs HF-Sh), TNF (5 ×; vs C-Sp and 1.7 ×;vs HF-Sh), IL-17 (1.7 ×; vs C-Sp), IL-10 (7.4 ×; vs C-Sp and 1.3 ×; vs HF-Sh), and modulated lipid metabolism in septic mice. In the HF-Sp group liver's, we observed hepatomegaly, hydropic degeneration, necrosis, an increase in oxidative stress (reduction of CAT activity (-81.7%; vs HF-Sh); increase MDA levels (82.8%; vs HF-Sh), and hepatic IL-6 (1.9 ×; vs HF-Sh), and TNF (1.3 × %;vs HF-Sh) production. Furthermore, we found a decrease in the total number of inflammatory, mononuclear cells, and in the regenerative processes, and binucleated hepatocytes in a HF-Sp group liver's. Our results suggested that the organism under metabolic stress of a HF diet during sepsis may worsen the inflammatory landscape and hepatocellular injury and may harm the liver regenerative process.
Collapse
Affiliation(s)
- Sttefany Viana Gomes
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Bruna Vidal Dias
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Pedro Alves Machado Júnior
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Renata Rebeca Pereira
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Débora Maria Soares de Souza
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Gustavo Silveira Breguez
- Multiuser Research Laboratory, School of Nutrition, School of Nutrition, Postgraduate Program in Health and Nutrition, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Wanderson Geraldo de Lima
- Morphopathology Laboratory, Department of Biological Sciences (DECBI), Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Cintia Lopes de Brito Magalhães
- Laboratory of Biology and Technology of Microorganisms (LBTM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Silvia Dantas Cangussú
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Karina Barbosa Queiroz
- Laboratory of Experimental Nutrition (LABNEx), Department of Food, Postgraduate Program in Health and Nutrition, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Allan Jefferson Cruz Calsavara
- Laboratory of Cognition and Health (LACOS), School of Medicine, Department of Pediatric and Adult Clinics (DECPA), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Daniela Caldeira Costa
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil.
| |
Collapse
|
8
|
Barthelemy J, Bogard G, Wolowczuk I. Beyond energy balance regulation: The underestimated role of adipose tissues in host defense against pathogens. Front Immunol 2023; 14:1083191. [PMID: 36936928 PMCID: PMC10019896 DOI: 10.3389/fimmu.2023.1083191] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/09/2023] [Indexed: 03/06/2023] Open
Abstract
Although the adipose tissue (AT) is a central metabolic organ in the regulation of whole-body energy homeostasis, it is also an important endocrine and immunological organ. As an endocrine organ, AT secretes a variety of bioactive peptides known as adipokines - some of which have inflammatory and immunoregulatory properties. As an immunological organ, AT contains a broad spectrum of innate and adaptive immune cells that have mostly been studied in the context of obesity. However, overwhelming evidence supports the notion that AT is a genuine immunological effector site, which contains all cell subsets required to induce and generate specific and effective immune responses against pathogens. Indeed, AT was reported to be an immune reservoir in the host's response to infection, and a site of parasitic, bacterial and viral infections. In addition, besides AT's immune cells, preadipocytes and adipocytes were shown to express innate immune receptors, and adipocytes were reported as antigen-presenting cells to regulate T-cell-mediated adaptive immunity. Here we review the current knowledge on the role of AT and AT's immune system in host defense against pathogens. First, we will summarize the main characteristics of AT: type, distribution, function, and extraordinary plasticity. Second, we will describe the intimate contact AT has with lymph nodes and vessels, and AT immune cell composition. Finally, we will present a comprehensive and up-to-date overview of the current research on the contribution of AT to host defense against pathogens, including the respiratory viruses influenza and SARS-CoV-2.
Collapse
Affiliation(s)
| | | | - Isabelle Wolowczuk
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019 - UMR 9017 - Center for Infection and Immunity of Lille (CIIL), Lille, France
| |
Collapse
|
9
|
Yin Y, Xie Y, Ge W, Li Y. Creeping fat formation and interaction with intestinal disease in Crohn's disease. United European Gastroenterol J 2022; 10:1077-1084. [PMID: 36507842 PMCID: PMC9752293 DOI: 10.1002/ueg2.12349] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
Creeping fat (CrF), also known as fat wrapping, is a significant disease characteristic of Crohn's disease (CD). The transmural inflammation impairs intestinal integrity and facilitates bacteria translocation, aggravating immune response. CrF is a rich source of pro-inflammatory and pro-fibrotic cytokines with complex immune microenvironment. The inflamed and stricturing intestine is often wrapped by CrF, and CrF is associated with greater severity of CD. The large amount of innate and adaptive immune cells as well as adipocytes in CrF promote fibrosis in the affected intestine by secreting large amount of pro-fibrotic cytokines, adipokines, growth factors and fatty acids. CrF is a potential therapeutic target for CD treatment and a promising bio-marker for predicting response to drug therapy. This review aims to summarize and update the clinical manifestation and application of CrF and the underlying molecular mechanism involved in the pathogenesis of intestinal inflammation and fibrosis in CD.
Collapse
Affiliation(s)
- Yi Yin
- Department of General SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Ying Xie
- Department of General SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Wei Ge
- Department of General SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Yi Li
- Department of General SurgeryJinling HospitalMedical School of Nanjing UniversityNanjingChina
| |
Collapse
|
10
|
Campos-Sánchez JC, Gonzalez-Silvera D, Gong X, Broughton R, Guardiola FA, Betancor MB, Esteban MÁ. Implication of adipocytes from subcutaneous adipose tissue and fatty acids in skin inflammation caused by λ-carrageenin in gilthead seabream (Sparusaurata). FISH & SHELLFISH IMMUNOLOGY 2022; 131:160-171. [PMID: 36210005 DOI: 10.1016/j.fsi.2022.09.066] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
The role of subcutaneous adipose tissue adipocytes and the effects of fatty acids on carrageenan-induced skin inflammation in gilthead seabream (Sparus aurata) were studied. Fish were injected intramuscularly with phosphate-buffered saline (control) or λ-carrageenin (1%), and skin samples collected at the injection site at 3 and 6 h post-injection (p.i.) were processed for histological study. In addition, the presence and levels of lipid classes, fatty acid methyl esters (FAME) and eicosanoids were evaluated in the skin samples obtained from the injected areas. Histological results indicated an increase in adipocyte area in fish sampled at 3 h p.i. with λ-carrageenin compared to fish in the control group. Furthermore, the frequency of adipocytes between 4500 and 5000 μm2 was increased at 6 h in the λ-carrageenin group compared to the control group. Analysis of lipid classes found that fish injected with λ-carrageenan showed increased free fatty acid (FFA) and sphingomyelin content at 3 and 6 h, respectively, compared to the control group. An increase in saturated fatty acids (SFA), n-6 polyunsaturated fatty acids (PUFA), and a decrease in the values of monounsaturated fatty acids (MUFA), n-3 PUFA and minor fatty acids were observed in fish skin at 6 h after λ-carrageenin injection, with respect to the values obtained in the control group. Regarding the analysis of eicosanoids, an increase in hydroxyeicosatetraenoic acid (5-HETE) was detected in the skin of fish at 6 h post-carrageenin injection compared to the control group. The presented results indicate the contribution of adipocytes and fatty acids in the development and regulation of the inflammatory response triggered by λ-carrageenin in gilthead seabream skin.
Collapse
Affiliation(s)
- Jose Carlos Campos-Sánchez
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology. Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - Daniel Gonzalez-Silvera
- Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, Scotland, UK
| | - Xu Gong
- Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, Scotland, UK
| | - Richard Broughton
- Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, Scotland, UK
| | - Francisco A Guardiola
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology. Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - Mónica B Betancor
- Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, Scotland, UK
| | - María Ángeles Esteban
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology. Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain.
| |
Collapse
|
11
|
Michalak A, Kasztelan-Szczerbińska B, Cichoż-Lach H. Impact of Obesity on the Course of Management of Inflammatory Bowel Disease—A Review. Nutrients 2022; 14:nu14193983. [PMID: 36235636 PMCID: PMC9573343 DOI: 10.3390/nu14193983] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
It is already well-known that visceral adipose tissue is inseparably related to the pathogenesis, activity, and general outcome of inflammatory bowel disease (IBD). We are getting closer and closer to the molecular background of this loop, finding certain relationships between activated mesenteric tissue and inflammation within the lumen of the gastrointestinal tract. Recently, relatively new data have been uncovered, indicating a direct impact of body fat on the pattern of pharmacological treatment in the course of IBD. On the other hand, ileal and colonic types of Crohn’s disease and ulcerative colitis appear to be more diversified than it was thought in the past. However, the question arises whether at this stage we are able to translate this knowledge into the practical management of IBD patients or we are still exploring the scientific background of this pathology, having no specific tools to be used directly in patients. Our review explores IBD in the context of obesity and associated disorders, focusing on adipokines, creeping fat, and possible relationships between these disorders and the treatment of IBD patients.
Collapse
|
12
|
Suau R, Pardina E, Domènech E, Lorén V, Manyé J. The Complex Relationship Between Microbiota, Immune Response and Creeping Fat in Crohn's Disease. J Crohns Colitis 2022; 16:472-489. [PMID: 34528668 DOI: 10.1093/ecco-jcc/jjab159] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the last decade, there has been growing interest in the pathological involvement of hypertrophic mesenteric fat attached to the serosa of the inflamed intestinal segments involved in Crohn's disease [CD], known as creeping fat. In spite of its protective nature, creeping fat harbours an aberrant inflammatory activity which, in an already inflamed intestine, may explain why creeping fat is associated with a greater severity of CD. The transmural inflammation of CD facilitates the interaction of mesenteric fat with translocated intestinal microorganisms, contributing to activation of the immune response. This may be not the only way in which microorganisms alter the homeostasis of this fatty tissue: intestinal dysbiosis may also impair xenobiotic metabolism. All these CD-related alterations have a functional impact on nuclear receptors such as the farnesoid X receptor or the peroxisome proliferator-activated receptor γ, which are implicated in regulation of the immune response, adipogenesis and the maintenance of barrier function, as well as on creeping fat production of inflammatory-associated cells such as adipokines. The dysfunction of creeping fat worsens the inflammatory course of CD and may favour intestinal fibrosis and fistulizing complications. However, our current knowledge of the pathophysiology and pathogenic role of creeping fat is controversial and a better understanding might provide new therapeutic targets for CD. Here we aim to review and update the key cellular and molecular alterations involved in this inflammatory process that link the pathological components of CD with the development of creeping fat.
Collapse
Affiliation(s)
- Roger Suau
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| | - Eva Pardina
- Biochemistry and Molecular Biomedicine Department, University of Barcelona, Barcelona (Catalonia), Spain
| | - Eugeni Domènech
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain.,Gastroenterology Department, 'Germans Trias i Pujol' University Hospital, Badalona (Catalonia), Spain
| | - Violeta Lorén
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| | - Josep Manyé
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| |
Collapse
|
13
|
Abstract
Two decades of research have established that Nuclear Factor-κB (NF-κB) signaling plays a critical role in reprogramming the fat cell transcriptome towards inflammation in response to overnutrition and metabolic stress. Several groups have suggested that inhibition of NF-κB signaling could have metabolic benefits for obesity-associated adipose tissue inflammation. However, two significant problems arise with this approach. The first is how to deliver general NF-κB inhibitors into adipocytes without allowing these compounds to disrupt normal functioning in cells of the immune system. The second issue is that general inhibition of canonical NF-κB signaling in adipocytes will likely lead to a massive increase in adipocyte apoptosis under conditions of metabolic stress, leading full circle into a secondary inflammation (However, this problem may not be true for non-canonical NF-κB signaling.). This review will focus on the research that has examined canonical and non-canonical NF-κB signaling in adipocytes, focusing on genetic studies that examine loss-of-function of NF-κB specifically in fat cells. Although the development of general inhibitors of canonical NF-κB signaling seems unlikely to succeed in alleviating adipose tissue inflammation in humans, the door remains open for more targeted therapeutics. In principle, these would include compounds that interrogate NF-κB DNA binding, protein-protein interactions, or post-translational modifications that partition NF-κB activity towards some genes and away from others in adipocytes. I also discuss the possibility for inhibitors of non-canonical NF-κB signaling to realize success in mitigating fat cell dysfunction in obesity. To plant the seeds for such approaches, much biochemical “digging” in adipocytes remains; this includes identifying—in an unbiased manner–NF-κB direct and indirect targets, genomic DNA binding sites for all five NF-κB subunits, NF-κB protein-protein interactions, and post-translational modifications of NF-κB in fat cells.
Collapse
|
14
|
Yin Y, Zhu ZX, Li Z, Chen YS, Zhu WM. Role of mesenteric component in Crohn’s disease: A friend or foe? World J Gastrointest Surg 2021; 13:1536-1549. [PMID: 35070062 PMCID: PMC8727179 DOI: 10.4240/wjgs.v13.i12.1536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 08/01/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023] Open
Abstract
Crohn’s disease (CD) is a complex and relapsing gastrointestinal disease with mesenteric alterations. The mesenteric neural, vascular, and endocrine systems actively take part in the gut dysbiosis-adaptive immunity-mesentery-body axis, and this axis has been proven to be bidirectional. The abnormalities of morphology and function of the mesenteric component are associated with intestinal inflammation and disease progress of CD via responses to afferent signals, neuropeptides, lymphatic drainage, adipokines, and functional cytokines. The hypertrophy of mesenteric adipose tissue plays important roles in the pathogenesis of CD by secreting large amounts of adipokines and representing a rich source of proinflammatory or profibrotic cytokines. The vascular alteration, including angiogenesis and lymphangiogenesis, is concomitant in the disease course of CD. Of note, the enlarged and obstructed lymphatic vessels, which have been described in CD patients, are likely related to the early onset submucosa edema and being a cause of CD. The function of mesenteric lymphatics is influenced by endocrine of mesenteric nerves and adipocytes. Meanwhile, the structure of the mesenteric lymphatic vessels in hypertrophic mesenteric adipose tissue is mispatterned and ruptured, which can lead to lymph leakage. Leaky lymph factors can in turn stimulate adipose tissue to proliferate and effectively elicit an immune response. The identification of the role of mesentery and the crosstalk between mesenteric tissues in intestinal inflammation may shed light on understanding the underlying mechanism of CD and help explore new therapeutic targets.
Collapse
Affiliation(s)
- Yi Yin
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Zhen-Xing Zhu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Zhun Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Yu-Sheng Chen
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Wei-Ming Zhu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| |
Collapse
|
15
|
de Jesus JDCR, Murari ASDP, Radloff K, de Moraes RCM, Figuerêdo RG, Pessoa AFM, Rosa-Neto JC, Matos-Neto EM, Alcântara PSM, Tokeshi F, Maximiano LF, Bin FC, Formiga FB, Otoch JP, Seelaender M. Activation of the Adipose Tissue NLRP3 Inflammasome Pathway in Cancer Cachexia. Front Immunol 2021; 12:729182. [PMID: 34630405 PMCID: PMC8495409 DOI: 10.3389/fimmu.2021.729182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/03/2021] [Indexed: 12/11/2022] Open
Abstract
Background Cachexia is a paraneoplastic syndrome that accompanies and compromises cancer treatment, especially in advanced stages, affecting the metabolism and function of several organs. The adipose tissue is the first to respond to the presence of the tumor, contributing to the secretion of factors which drive the systemic inflammation, a hallmark of the syndrome. While inflammation is a defensive innate response, the control mechanisms have been reported to be disrupted in cachexia. On the other hand, little is known about the role of NLRP3 inflammasome in this scenario, a multiprotein complex involved in caspase-1 activation and the processing of the cytokines IL-1β and IL-18. Aim based on the evidence from our previous study with a rodent model of cachexia, we examined the activation of the NLRP3 inflammasome pathway in two adipose tissue depots obtained from patients with colorectal cancer and compared with that another inflammatory pathway, NF-κB. Results For CC we found opposite modulation in ScAT and PtAT for the gene expression of TLR4, Caspase-1 (cachectic group) and for NF-κB p50, NF-κB p65, IL-1β. CD36, expression was decreased in both depots while that of NLRP3 and IL-18 was higher in both tissues, as compared with controls and weight stable patients (WSC). Caspase-1 basal protein levels in the ScAT culture supernatant were higher in WSC and (weight stable patients) CC, when compared to controls. Basal ScAT explant culture medium IL-1β and IL-18 protein content in ScAT supernatant was decreased in the WSC and CC as compared to CTL explants. Conclusions The results demonstrate heterogeneous responses in the activation of genes of the NLRP3 inflammasome pathway in the adipose tissue of patients with cancer cachexia, rendering this pathway a potential target for therapy aiming at decreasing chronic inflammation in cancer.
Collapse
Affiliation(s)
- Joyce de Cassia Rosa de Jesus
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ariene Soares de Pinho Murari
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Katrin Radloff
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ruan Carlos Macêdo de Moraes
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Raquel Galvão Figuerêdo
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Flavia Marçal Pessoa
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - José César Rosa-Neto
- Immunometabolism Laboratory, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Emídio Marques Matos-Neto
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Paulo S M Alcântara
- University Hospital, Department of Surgical Clinic, Universidade de São Paulo, São Paulo, Brazil
| | - Flavio Tokeshi
- University Hospital, Department of Surgical Clinic, Universidade de São Paulo, São Paulo, Brazil
| | - Linda Ferreira Maximiano
- University Hospital, Department of Surgical Clinic, Universidade de São Paulo, São Paulo, Brazil
| | - Fang Chia Bin
- Department of Coloproctology, Santa Casa de São Paulo, São Paulo, Brazil
| | | | - José P Otoch
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,University Hospital, Department of Surgical Clinic, Universidade de São Paulo, São Paulo, Brazil
| | - Marilia Seelaender
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
Karrasch T, Höpfinger A, Schäffler A, Schmid A. The adipokine C1q/TNF-related protein-3 (CTRP-3) inhibits Toll-like receptor (TLR)-induced expression of Cathelicidin antimicrobial peptide (CAMP) in adipocytes. Cytokine 2021; 148:155663. [PMID: 34388476 DOI: 10.1016/j.cyto.2021.155663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIM CAMP (Cathelicidin antimicrobial peptide) expression in adipocytes is regulated by Toll-like receptor (TLR) agonists. Secreted adipokines such as CTRP-3 have been suggested to participate in innate immune signaling in adipose tissue (AT). This study investigates whether TLR-induced CAMP expression in adipocytes is antagonized by CTRP-3. METHODS 3T3-L1 adipocytes were co-stimulated with TLR agonists (LPS, MALP-2, Pam3CSK4, pI:C) and recombinant CTRP-3. In a SIRS model, C57BL/6 wild-type mice were intraperitoneally (ip) injected with recombinant CTRP-3 prior to LPS. CAMP expression was analyzed by real-time PCR in AT of wild-type mice and in AT and primary adipocytes from transgenic mice lacking adipocyte CTRP-3 expression. Comparative transcriptome analysis by RNA seq. was applied in CTRP-3 KO adipocytes. RESULTS In vitro, CTRP-3 antagonized TLR4- and TLR1/2-induced CAMP expression in adipocytes whereas TLR3- and TLR2/6-mediated induction of CAMP was not affected. in vivo, application of exogenous CTRP-3 dose-dependently antagonized LPS-induced CAMP expression in intra-abdominal AT. CAMP expression in total AT and in primary adipocytes of subcutaneous and intra-abdominal AT did not differ between wild-type mice and transgenic mice lacking adipocyte CTRP-3 expression. CONCLUSIONS The study suggests a hypothetical role of CAMP in host defense not only against Gram-positive bacteria sensed by TLR1/2 and TLR2/6 but also against Gram-negative bacteria sensed by TLR4 and potentially against viruses sensed by TLR3. The machinery of TLR-mediated pro-inflammatory activation of the CAMP gene in adipocytes seems to be partly modulated by secreted adipokines belonging to the growing family of C1q/TNF-related proteins such as CTRP-3.
Collapse
Affiliation(s)
- Thomas Karrasch
- Department of Internal Medicine III, University of Giessen, Germany
| | | | | | - Andreas Schmid
- Department of Internal Medicine III, University of Giessen, Germany.
| |
Collapse
|
17
|
Danforth DN. The Role of Chronic Inflammation in the Development of Breast Cancer. Cancers (Basel) 2021; 13:3918. [PMID: 34359821 PMCID: PMC8345713 DOI: 10.3390/cancers13153918] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/20/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic inflammation contributes to the malignant transformation of several malignancies and is an important component of breast cancer. The role of chronic inflammation in the initiation and development of breast cancer from normal breast tissue, however, is unclear and needs to be clarified. A review of the literature was conducted to define the chronic inflammatory processes in normal breast tissue at risk for breast cancer and in breast cancer, including the role of lymphocyte and macrophage infiltrates, chronic active adipocytes and fibroblasts, and processes that may promote chronic inflammation including the microbiome and factors related to genomic abnormalities and cellular injury. The findings indicate that in healthy normal breast tissue there is systemic evidence to suggest inflammatory changes are present and associated with breast cancer risk, and adipocytes and crown-like structures in normal breast tissue may be associated with chronic inflammatory changes. The microbiome, genomic abnormalities, and cellular changes are present in healthy normal breast tissue, with the potential to elicit inflammatory changes, while infiltrating lymphocytes are uncommon in these tissues. Chronic inflammatory changes occur prominently in breast cancer tissues, with important contributions from tumor-infiltrating lymphocytes and tumor-associated macrophages, cancer-associated adipocytes and crown-like structures, and cancer-associated fibroblasts, while the microbiome and DNA damage may serve to promote inflammatory events. Together, these findings suggest that chronic inflammation may play a role in influencing the initiation, development and conduct of breast cancer, although several chronic inflammatory processes in breast tissue may occur later in breast carcinogenesis.
Collapse
Affiliation(s)
- David N Danforth
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Schmid A, Schäffler A, Karrasch T. CTRP-3 Regulates NOD1-mediated Inflammation and NOD1 Expression in Adipocytes and Adipose Tissue. Inflammation 2021; 44:2260-2269. [PMID: 34165676 PMCID: PMC8616866 DOI: 10.1007/s10753-021-01497-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 11/29/2022]
Abstract
The anti-inflammatory adipokine CTRP-3 might affect innate immune reactions such as NOD1. The impact of CTRP-3 on NOD1-mediated inflammation in adipocytes and monocytic cells as well as on NOD1 expression was investigated. Murine 3T3-L1 pre-adipocytes and adipocytes as well as human THP-1 monocyte-like cells were co-stimulated with the synthetic NOD1 agonist Tri-DAP and recombinant CTRP-3. Gonadal adipose tissue and primary adipocytes were obtained from a murine model carrying a knockout (KO) of CTRP-3 in adipocytes but not in stroma-vascular cells. Wildtype mice with lipopolysaccharide (LPS)-induced elevated NOD1 expression were treated with CTRP-3. Secreted inflammatory cytokines in cell supernatants were measured by ELISA and mRNA levels were quantified by RT-PCR. Pro-inflammatory chemokine and cytokine secretion (MCP-1, RANTES, TNFα) was induced by NOD1 activation in adipocytes and monocyte-like cells, and MCP-1 and RANTES release was effectively inhibited by pre-incubation of cells with CTRP-3. CTRP-3 also antagonized LPS-triggered induction of NOD1 gene expression in murine adipose tissue, whereas adipocyte CTRP-3 deficiency upregulated NOD1 expression in adipose tissue. CTRP-3 is an effective antagonist of peptidoglycan-induced, NOD1-mediated inflammation and of LPS-induced NOD1 expression. Since basal NOD1 expression is increased by adipocyte CTRP-3 deficiency, there have to be also inflammation-independent mechanisms of NOD1 expression regulation by CTRP-3.
Collapse
Affiliation(s)
- Andreas Schmid
- Department of Internal Medicine III, University Hospital of Giessen, Giessen, Germany.
| | - Andreas Schäffler
- Department of Internal Medicine III, University Hospital of Giessen, Giessen, Germany
| | - Thomas Karrasch
- Department of Internal Medicine III, University Hospital of Giessen, Giessen, Germany
| |
Collapse
|
19
|
Höpfinger A, Karrasch T, Schäffler A, Schmid A. Regulation of CAMP (cathelicidin antimicrobial peptide) expression in adipocytes by TLR 2 and 4. Innate Immun 2021; 27:184-191. [PMID: 33509002 PMCID: PMC7882808 DOI: 10.1177/1753425920988167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Recent data argue for a pro-inflammatory role of CAMP (cathelicidin antimicrobial peptide) in adipocytes and adipose tissue (AT) and for regulatory circuits involving TLRs. In order to investigate regulatory effects of TLR2 and TLR4, 3T3-L1 adipocytes were stimulated with TLR2 agonistic lipopeptide MALP-2 and with TLR4 agonist LPS in presence or absence of signal transduction inhibitors. CAMP gene expression was analysed by quantitative real-time PCR in adipocytes and in murine AT compartments and cellular subfractions. CAMP expression was higher in gonadal than in subcutaneous AT and there was a gender-specific effect with higher levels in males. Adipocytes had higher CAMP expression than the stroma-vascular cell (SVC) fraction. MALP-2 up-regulated CAMP expression significantly, mediated by STAT3 and PI3K and potentially (non-significant trend) by NF-κB and MAPK, but not by raf-activated MEK-1/-2. Moreover, LPS proved to act as a potent inducer of CAMP via NF-κB, PI3K and STAT3, whereas specific inhibition of MAPK and MEK-1/-2 had no effect. In conclusion, activation of TLR2 and TLR4 by classical ligands up-regulates adipocyte CAMP expression involving classical signal transduction elements. These might represent future drug targets for pharmacological modulation of CAMP expression in adipocytes, especially in the context of metabolic and infectious diseases.
Collapse
Affiliation(s)
| | - Thomas Karrasch
- Department of Internal Medicine III, University of Giessen, Germany
| | | | - Andreas Schmid
- Department of Internal Medicine III, University of Giessen, Germany
| |
Collapse
|
20
|
Evaluation of Fat Accumulation and Adipokine Production during the Long-Term Adipogenic Differentiation of Porcine Intramuscular Preadipocytes and Study of the Influence of Immunobiotics. Cells 2020; 9:cells9071715. [PMID: 32708964 PMCID: PMC7408200 DOI: 10.3390/cells9071715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/17/2022] Open
Abstract
The degree of fat accumulation and adipokine production are two major indicators of obesity that are correlated with increased adipose tissue mass and chronic inflammatory responses. Adipocytes have been considered effector cells for the inflammatory responses due to their capacity to express Toll-like receptors (TLRs). In this study, we evaluated the degree of fat accumulation and adipokine production in porcine intramuscular preadipocyte (PIP) cells maintained for in vitro differentiation over a long period without or with stimulation of either TNF-α or TLR2-, TLR3-, or TLR4-ligands. The cytosolic fat accumulation was measured by liquid chromatography and the expression of adipokines (CCL2, IL-6, IL-8 and IL-10) were quantified by RT-qPCR and ELISA at several time points (0 to 20 days) of PIP cells differentiation. Long-term adipogenic differentiation (LTAD) induced a progressive fat accumulation in the adipocytes over time. Activation of TLR3 and TLR4 resulted in an increased rate of fat accumulation into the adipocytes over the LTAD. The production of CCL2, IL-8 and IL-6 were significantly increased in unstimulated adipocytes during the LTAD, while IL-10 expression remained stable over the studied period. An increasing trend of adiponectin and leptin production was also observed during the LTAD. On the other hand, the stimulation of adipocytes with TLRs agonists or TNF-α resulted in an increasing trend of CCL2, IL-6 and IL-8 production while IL-10 remained stable in all four treatments during the LTAD. We also examined the influences of several immunoregulatory probiotic strains (immunobiotics) on the modulation of the fat accumulation and adipokine production using supernatants of immunobiotic-treated intestinal immune cells and the LTAD of PIP cells. Immunobiotics have shown a strain-specific ability to modulate the fat accumulation and adipokine production, and differentiation of adipocytes. Here, we expanded the utility and potential application of our in vitro PIP cells model by evaluating an LTAD period (20 days) in order to elucidate further insights of chronic inflammatory pathobiology of adipocytes associated with obesity as well as to explore the prospects of immunomodulatory intervention for obesity such as immunobiotics.
Collapse
|
21
|
Abstract
Cardiovascular disease and infections are major causes for the high incidence of morbidity and mortality of patients with chronic kidney disease. Both complications are directly or indirectly associated with disturbed functions or altered apoptotic rates of polymorphonuclear leukocytes, monocytes, lymphocytes, and dendritic cells. Normal responses of immune cells can be reduced, leading to infectious diseases or pre-activated/primed, giving rise to inflammation and subsequently to cardiovascular disease. This review summarizes the impact of kidney dysfunction on the immune system. Renal failure results in disturbed renal metabolic activities with reduced renin, erythropoietin, and vitamin D production, which adversely affects the immune system. Decreased kidney function also leads to reduced glomerular filtration and the retention of uremic toxins. A large number of uremic toxins with detrimental effects on immune cells have been identified. Besides small water-soluble and protein-bound compounds originating from the intestinal microbiome, several molecules in the middle molecular range, e.g., immunoglobulin light chains, retinol-binding protein, the neuropeptides Met-enkephalin and neuropeptide Y, endothelin-1, and the adipokines leptin and resistin, adversely affect immune cells. Posttranslational modifications such as carbamoylation, advanced glycation products, and oxidative modifications contribute to uremic toxicity. Furthermore, high-density lipoprotein from uremic patients has an altered protein profile and thereby loses its anti-inflammatory properties.
Collapse
Affiliation(s)
- Gerald Cohen
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna A-1090, Austria
| |
Collapse
|
22
|
Chirumbolo S. Oxidative Stress, Nutrition and Cancer: Friends or Foes? World J Mens Health 2020; 39:19-30. [PMID: 32202081 PMCID: PMC7752511 DOI: 10.5534/wjmh.190167] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022] Open
Abstract
The relationship between cancer and nutrition, as well as nutrition and oxidative stress, shares puzzling aspects that current research is investigating as the possible components of an intriguing regulating mechanism involving the complex interplay between adipose tissue and other compartments. Along the very recent biological evolution, humans underwent a rapid change in their lifestyles and henceforth the role of the adipocytes earned a much more complex task in the fine tuning of the tissue microenvironment. A lipidic signaling language probably evolved in association with the signaling role of reactive oxygen species, which gained a fundamental part in the regulation of cell stem and plasticity. The possible relationship with cancer onset might have some causative mechanism in the impairment of this complex task, usually deregulated by drastic changes in one's own lifestyle and dietary habit. This review tries to address this issue.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| |
Collapse
|
23
|
Oates JR, McKell MC, Moreno-Fernandez ME, Damen MSMA, Deepe GS, Qualls JE, Divanovic S. Macrophage Function in the Pathogenesis of Non-alcoholic Fatty Liver Disease: The Mac Attack. Front Immunol 2019; 10:2893. [PMID: 31921154 PMCID: PMC6922022 DOI: 10.3389/fimmu.2019.02893] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 11/26/2019] [Indexed: 12/15/2022] Open
Abstract
Obesity is a prevalent predisposing factor to non-alcoholic fatty liver disease (NAFLD), the most common chronic liver disease in the developed world. NAFLD spectrum of disease involves progression from steatosis (NAFL), to steatohepatitis (NASH), cirrhosis and hepatocellular carcinoma (HCC). Despite clinical and public health significance, current FDA approved therapies for NAFLD are lacking in part due to insufficient understanding of pathogenic mechanisms driving disease progression. The etiology of NAFLD is multifactorial. The induction of both systemic and tissue inflammation consequential of skewed immune cell metabolic state, polarization, tissue recruitment, and activation are central to NAFLD progression. Here, we review the current understanding of the above stated cellular and molecular processes that govern macrophage contribution to NAFLD pathogenesis and how adipose tissue and liver crosstalk modulates macrophage function. Notably, the manipulation of such events may lead to the development of new therapies for NAFLD.
Collapse
Affiliation(s)
- Jarren R Oates
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Melanie C McKell
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Michelle S M A Damen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - George S Deepe
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Joseph E Qualls
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
24
|
Liddle DM, Monk JM, Hutchinson AL, Ma DWL, Robinson LE. CD8 + T cell/adipocyte inflammatory cross talk and ensuing M1 macrophage polarization are reduced by fish-oil-derived n-3 polyunsaturated fatty acids, in part by a TNF-α-dependent mechanism. J Nutr Biochem 2019; 76:108243. [PMID: 31760229 DOI: 10.1016/j.jnutbio.2019.108243] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/15/2019] [Accepted: 09/10/2019] [Indexed: 12/25/2022]
Abstract
Obese visceral adipose tissue (AT) inflammation is driven by adipokine-mediated cross talk between CD8+ T cells and adipocytes, a process mitigated by long-chain (LC) n-3 polyunsaturated fatty acids (PUFA) but underlying mechanisms and ensuing effects on macrophage polarization status are unknown. Using an in vitro co-culture model that recapitulates the degree of CD8+ T cell infiltration reported in obese AT, 3T3-L1 adipocytes were co-cultured for 24 h with purified splenic CD8+ T cells from C57Bl/6 mice consuming either a 10% w/w safflower oil (control, CON) or 7% w/w safflower oil + 3% w/w fish oil (FO) diet for 4 weeks (n=8-10/diet). Co-cultured cells were in direct contact or in a contact-independent condition separated by a Transwell permeable membrane and stimulated with lipopolysaccharide (10 ng/ml) to mimic in vivo obese endotoxin levels. In contact-dependent co-cultures, FO reduced inflammatory (IL-6, TNFα, IFN-γ) and macrophage chemotactic (CCL2, CCL7, CCL3) mRNA expression and/or secreted protein, NF-κB p65 activation, ROS accumulation, NLRP3 inflammasome priming (Nlrp3, Il1β mRNA) and activation (caspase-1 activity) compared to CON (P<.05). The anti-inflammatory action of FO was reproduced by the addition of a TNF-α neutralizing antibody (1 μg/ml) to CON co-cultures (CON/anti-TNF-α), albeit to a lesser degree. Conditioned media from FO and CON/anti-TNF-α co-cultures, in turn, reduced RAW 264.7 macrophage mRNA expression of M1 polarization markers (iNos, Cd11c, Ccr2) and associated inflammatory cytokines (Il6, Tnfα, Il1β) compared to CON. These data suggest that inflammatory CD8+ T cell/adipocyte cross talk is partially attributable to TNF-α signaling, which can be mitigated by LC n-3 PUFA.
Collapse
Affiliation(s)
- Danyelle M Liddle
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Jennifer M Monk
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Amber L Hutchinson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - David W L Ma
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Lindsay E Robinson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1.
| |
Collapse
|
25
|
Fish oil supplementation to a high-fat diet improves both intestinal health and the systemic obese phenotype. J Nutr Biochem 2019; 72:108216. [DOI: 10.1016/j.jnutbio.2019.07.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 06/28/2019] [Accepted: 07/19/2019] [Indexed: 12/25/2022]
|
26
|
The Role of Adipose Tissue in the Pathogenesis and Therapeutic Outcomes of Inflammatory Bowel Disease. Cells 2019; 8:cells8060628. [PMID: 31234447 PMCID: PMC6627060 DOI: 10.3390/cells8060628] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/16/2019] [Accepted: 06/18/2019] [Indexed: 02/06/2023] Open
Abstract
Though historically regarded as an inert energy store, adipose tissue is a complex endocrine organ, which is increasingly implicated in the pathogenesis of inflammatory bowel disease (IBD). Accumulating evidence points to visceral adipose tissue and specifically to its mesenteric component, or “creeping fat” as impacting on the disease course through its immunomodulatory properties. On the one hand, mesenteric fat acts as a physical barrier to inflammation and is involved in controlling host immune response to translocation of gut bacteria. On the other hand, however, there exists a strong link between visceral fat and complicated course of the disease with unfavorable therapeutic outcomes. Furthermore, “creeping fat” appears to play different roles in different IBD phenotypes, with the greatest pathogenetic contribution probably to an ileal form of Crohn’s disease. In this review, we summarize and discuss the existing literature on the subject and identify high-priority areas for future research. It may be that a better understanding of the role of mesenteric fat in IBD will determine new therapeutic targets and translate into improved clinical outcomes.
Collapse
|
27
|
Igata M, Islam MA, Tada A, Takagi M, Kober AKMH, Albarracin L, Aso H, Ikeda-Ohtsubo W, Miyazawa K, Yoda K, He F, Takahashi H, Villena J, Kitazawa H. Transcriptome Modifications in Porcine Adipocytes via Toll-Like Receptors Activation. Front Immunol 2019; 10:1180. [PMID: 31191544 PMCID: PMC6549529 DOI: 10.3389/fimmu.2019.01180] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/09/2019] [Indexed: 12/15/2022] Open
Abstract
Adipocytes are the most important cell type in adipose tissue playing key roles in immunometabolism. We previously reported that nine members of the Toll-like receptor (TLR) family are expressed in an originally established porcine intramuscular pre-adipocyte (PPI) cell line. However, the ability of TLR ligands to modulate immunometabolic transcriptome modifications in porcine adipocytes has not been elucidated. Herein, we characterized the global transcriptome modifications in porcine intramuscular mature adipocytes (pMA), differentiated from PPI, following stimulation with Pam3csk4, Poly(I:C) or LPS which are ligands for TLR2, TLR3, and TLR4, respectively. Analysis of microarray data identified 530 (218 up, 312 down), 520 (245 up, 275 down), and 525 (239 up, 286 down) differentially expressed genes (DEGs) in pMA following the stimulation with Pam3csk4, Poly(I:C), and LPS, respectively. Gene ontology classification revealed that DEGs are involved in several biological processes including those belonging to immune response and lipid metabolism pathways. Functionally annotated genes were organized into two groups for downstream analysis: immune response related genes (cytokines, chemokines, complement factors, adhesion molecules, and signal transduction), and genes involved with metabolic and endocrine functions (hormones and receptors, growth factors, and lipid biosynthesis). Differential expression analysis revealed that EGR1, NOTCH1, NOS2, TNFAIP3, TRAF3IP1, INSR, CXCR4, PPARA, MAPK10, and C3 are the top 10 commonly altered genes of TLRs induced transcriptional modification of pMA. However, the protein-protein interaction network of DEGs identified EPOR, C3, STAR, CCL2, and SAA2 as the major hub genes, which were also exhibited higher centrality estimates in the Gene-Transcription factor interaction network. Our results provide new insights of transcriptome modifications associated with TLRs activation in porcine adipocytes and identified key regulatory genes that could be used as biomarkers for the evaluation of treatments having immunomodularoty and/or metabolic functional beneficial effects in porcine adipocytes.
Collapse
Affiliation(s)
- Manami Igata
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Md Aminul Islam
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Department of Medicine, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Asuka Tada
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Michihiro Takagi
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - A K M Humayun Kober
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Department of Dairy and Poultry Science, Chittagong Veterinary and Animal Sciences University, Chittangong, Bangladesh
| | - Leonardo Albarracin
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucumán, Argentina.,Scientific Computing Laboratory, Computer Science Department, Faculty of Exact Science and Technology, National University of Tucuman, San Miguel de Tucumán, Argentina
| | - Hisashi Aso
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Cell Biology Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Wakako Ikeda-Ohtsubo
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Kenji Miyazawa
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., Yokohama, Japan
| | - Kazutoyo Yoda
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., Yokohama, Japan
| | - Fang He
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., Yokohama, Japan
| | - Hideki Takahashi
- Laboratory of Plant Pathology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Plant Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Julio Villena
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucumán, Argentina
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
28
|
Chan CC, Damen MSMA, Alarcon PC, Sanchez-Gurmaches J, Divanovic S. Inflammation and Immunity: From an Adipocyte's Perspective. J Interferon Cytokine Res 2019; 39:459-471. [PMID: 30920343 DOI: 10.1089/jir.2019.0014] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Comprehension of adipocyte function has evolved beyond a long-held belief of their inert nature, as simple energy storing and releasing cells. Adipocytes, including white, brown, and beige, are capable mediators of global metabolic health, but their intersection with inflammation is a budding field of exploration. Evidence hints at a reciprocal relationship adipocytes share with immune cells. Adipocyte's capacity to behave in an "immune-like" manner and ability to sense inflammatory cues that subsequently alter core adipocyte function might play an important role in shaping immune responses. Clarifying this intricate relationship could uncover previously underappreciated contribution of adipocytes to inflammation-driven human health and disease. In this review, we highlight the potential of largely underappreciated adipocyte "immune-like" function and how it may contribute to inflammation, immunity, and pathology of various diseases.
Collapse
Affiliation(s)
- Calvin C Chan
- 1Medical Scientist Training Program, Immunology Graduate Program, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio.,2Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,3Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michelle S M A Damen
- 2Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,3Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Pablo C Alarcon
- 1Medical Scientist Training Program, Immunology Graduate Program, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio.,2Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,3Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joan Sanchez-Gurmaches
- 2Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,4Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,5Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Senad Divanovic
- 1Medical Scientist Training Program, Immunology Graduate Program, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio.,2Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,3Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,6Division of Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
29
|
Role of Dietary Lipids in Modulating Inflammation through the Gut Microbiota. Nutrients 2019; 11:nu11010117. [PMID: 30626117 PMCID: PMC6357048 DOI: 10.3390/nu11010117] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/19/2018] [Accepted: 12/30/2018] [Indexed: 12/12/2022] Open
Abstract
Inflammation and its resolution is a tenuous balance that is under constant contest. Though several regulatory mechanisms are employed to maintain homeostasis, disruptions in the regulation of inflammation can lead to detrimental effects for the host. Of note, the gut and microbial dysbiosis are implicated in the pathology of systemic chronic low-grade inflammation which has been linked to several metabolic diseases. What remains to be described is the extent to which dietary fat and concomitant changes in the gut microbiota contribute to, or arise from, the onset of metabolic disorders. The present review will highlight the role of microorganisms in host energy regulation and several mechanisms that contribute to inflammatory pathways. This review will also discuss the immunomodulatory effects of the endocannabinoid system and its link with the gut microbiota. Finally, a brief discussion arguing for improved taxonomic resolution (at the species and strain level) is needed to deepen our current knowledge of the microbiota and host inflammatory state.
Collapse
|
30
|
Adipose Tissue-Derived Biomarkers of Intestinal Barrier Functions for the Characterization of Diarrhoea-Predominant IBS. DISEASE MARKERS 2018; 2018:1827937. [PMID: 30622656 PMCID: PMC6304194 DOI: 10.1155/2018/1827937] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 12/17/2022]
Abstract
Background Alterations of the small-intestinal permeability (s-IP) might play an essential role in a subgroup of diarrhoea-predominant IBS (D-IBS) patients. Goals (a) To analyse in D-IBS patients the symptom profile in relation to the altered (+) or not (−) s-IP using the Gastrointestinal Symptom Rating Scale (GSRS). (b) To assess the circulating levels of the adipokines IL-6, IL-8, TNF-α, leptin, and adiponectin, along with LPS, TLR-4, neurotensin, and brain-derived neurotrophic factor (BDNF). The frequency distribution of SNPs at the loci for the investigated molecules and leptin receptor was evaluated. Study The study included 34 D-IBS patients and 17 healthy controls (HC). s-IP permeability was assayed by high-performance liquid chromatography determination in the urine of the lactulose to mannitol ratio. Concentrations of IL-6, IL-8, TNF-α, LPS, TLR-4, leptin, adiponectin, neurotensin, and BDNF were assayed by ELISA. Screening of genetic variants was done employing the restriction fragment length polymorphism-polymerase chain reaction method. Results D-IBS(−) patients had a significantly higher GSRS cluster pain and diarrhoea profile than D-IBS(+) ones. Significant correlations were found between the symptoms clusters and immune activation and inflammation markers. The levels of adipo(cyto)kines in D-IBS(+) patients were higher than those of controls, and IL-6 levels correlated with those of LPS. Leptin and BDNF were significantly higher, and neurotensin levels were significantly lower in D-IBS(+) than in controls. No differences were found in the frequency distribution of genotypes among the study groups. Conclusions Results from this study could be of some help in the characterization of the D-IBS and highlight the contribution of an altered intestinal barrier in the pathogenesis of this syndrome. Besides, a role could be ascribed to molecules secreted by the visceral adipose tissue that can impact on barrier functions.
Collapse
|
31
|
González FB, Villar SR, Toneatto J, Pacini MF, Márquez J, D’Attilio L, Bottasso OA, Piwien-Pilipuk G, Pérez AR. Immune response triggered by Trypanosoma cruzi infection strikes adipose tissue homeostasis altering lipid storage, enzyme profile and adipokine expression. Med Microbiol Immunol 2018; 208:651-666. [DOI: 10.1007/s00430-018-0572-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/30/2018] [Indexed: 12/11/2022]
|
32
|
Dokoshi T, Zhang LJ, Nakatsuji T, Adase CA, Sanford JA, Paladini RD, Tanaka H, Fujiya M, Gallo RL. Hyaluronidase inhibits reactive adipogenesis and inflammation of colon and skin. JCI Insight 2018; 3:123072. [PMID: 30385720 DOI: 10.1172/jci.insight.123072] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/13/2018] [Indexed: 12/16/2022] Open
Abstract
In this study we evaluated the role of hyaluronan (HA) in reactive adipogenesis, a local expansion of preadipocytes that provides host defense by release of antimicrobial peptides. We observed that HA accumulated during maturation of adipocytes in vitro and was associated with increased expression of preadipocyte factor 1, zinc finger protein 423, and early B cell factor 1. Although HA is normally abundant in the extracellular matrix, a further increase in HA staining occurred in mice at sites of reactive adipogenesis following injury of colon by dextran sodium sulfate or injury of skin from infection with Staphylococcus aureus. HA also abundantly accumulated around adipocytes seen in the colons of patients with inflammatory bowel disease. This HA was necessary for adipocyte maturation because digestion of HA by administration of soluble hyaluronidase or transgenic expression of hyaluronidase 1 inhibited adipogenesis in vitro and in vivo. Furthermore, hyaluronidase also suppressed inflammation of both skin and colon and decreased antimicrobial peptide expression by developing preadipocytes. This resulted in increased bacterial transit across the epithelial barrier despite decreased tissue injury from inflammation. These observations suggest HA plays an important role in reactive adipogenesis and host defense after injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hiroki Tanaka
- Department of Legal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Mikihiro Fujiya
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | | |
Collapse
|
33
|
Alwarawrah Y, Kiernan K, MacIver NJ. Changes in Nutritional Status Impact Immune Cell Metabolism and Function. Front Immunol 2018; 9:1055. [PMID: 29868016 PMCID: PMC5968375 DOI: 10.3389/fimmu.2018.01055] [Citation(s) in RCA: 293] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 04/27/2018] [Indexed: 12/15/2022] Open
Abstract
Immune cell function and metabolism are closely linked. Many studies have now clearly demonstrated that alterations in cellular metabolism influence immune cell function and that, conversely, immune cell function determines the cellular metabolic state. Less well understood, however, are the effects of systemic metabolism or whole organism nutritional status on immune cell function and metabolism. Several studies have demonstrated that undernutrition is associated with immunosuppression, which leads to both increased susceptibility to infection and protection against several types of autoimmune disease, whereas overnutrition is associated with low-grade, chronic inflammation that increases the risk of metabolic and cardiovascular disease, promotes autoreactivity, and disrupts protective immunity. Here, we review the effects of nutritional status on immunity and highlight the effects of nutrition on circulating cytokines and immune cell populations in both human studies and mouse models. As T cells are critical members of the immune system, which direct overall immune response, we will focus this review on the influence of systemic nutritional status on T cell metabolism and function. Several cytokines and hormones have been identified which mediate the effects of nutrition on T cell metabolism and function through the expression and action of key regulatory signaling proteins. Understanding how T cells are sensitive to both inadequate and overabundant nutrients may enhance our ability to target immune cell metabolism and alter immunity in both malnutrition and obesity.
Collapse
Affiliation(s)
- Yazan Alwarawrah
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Kaitlin Kiernan
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| | - Nancie J MacIver
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States.,Department of Immunology, Duke University Medical Center, Durham, NC, United States.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
34
|
Trejo-Vazquez F, Garza-Veloz I, Villela-Ramirez GA, Ortiz-Castro Y, Mauricio-Saucedo P, Cardenas-Vargas E, Diaz-Baez M, Cid-Baez MA, Castañeda-Miranda R, Ortiz-Rodriguez JM, Solis-Sanchez LO, Martinez-Fierro ML. Positive association between leptin serum levels and disease activity on endoscopy in inflammatory bowel disease: A case-control study. Exp Ther Med 2018; 15:3336-3344. [PMID: 29545852 PMCID: PMC5841033 DOI: 10.3892/etm.2018.5835] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/17/2017] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel disease (IBD) includes ulcerative colitis (UC), Crohn's disease (CD) and indeterminate colitis. As these subtypes of IBD display important differences in the behavior of the natural course of the disease, the identification of non-invasive markers for IBD is important. The aim of the present study was to evaluate the serum levels of 10 adipokines and their association with endoscopic activity in IBD. The 10-protein profile (C-peptide, ghrelin, gastric inhibitory polypeptide, glucagon-like peptide-1, glucagon, insulin, leptin, plasminogen activator inhibitor-1, resistin and visfatin) was evaluated using serum from 53 participants (23 UC and 11 CD patients, as well as 19 controls) from Zacatecas (Mexico) by using the Bio-Plex Pro Human Diabetes 10-Plex Panel (Bio-Rad Laboratories, Inc.). Compared with those in the controls, leptin levels were significantly lower in patients with IBD (P=4.9×10−4). In addition, serum leptin displayed differences between groups with and without disease activity on endoscopy (P<0.001). Among the study population, serum leptin levels of <5,494 pg/ml significantly increased the odds of IBD by 12.8-fold [odds ratio (OR)=12.8, 95% confidence interval (CI)=3.04–53.9, P=0.001]. In addition, patients with serum leptin levels of <2,498 pg/ml displayed 5.8-fold greater odds of disease activity on endoscopy among the study population (OR=5.8, 95% CI=1.52–22.4, P=0.013). No differences in the serum levels of the remaining proteins were identified between the groups. Among the study population, serum leptin was associated with an increased risk of IBD and with disease activity on endoscopy. Additional studies will be necessary to validate the use of leptin as a non-invasive biomarker of IBD severity.
Collapse
Affiliation(s)
- Fabiola Trejo-Vazquez
- Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud, Universidad Autonoma de Zacatecas 'Francisco García Salinas', Zacatecas 98160, Mexico.,Department of Gastroenterology, Hospital General de Zacatecas, Instituto de Seguridad y Servicios Sociales Para Los Trabajadores del Estado, Zacatecas 98000, Mexico
| | - Idalia Garza-Veloz
- Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud, Universidad Autonoma de Zacatecas 'Francisco García Salinas', Zacatecas 98160, Mexico.,Bioengineering Laboratory, Unidad Academica de Ingenieria Electrica, Universidad Autonoma de Zacatecas, Zacatecas 98000, Mexico
| | - Gabriela Alejandra Villela-Ramirez
- Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud, Universidad Autonoma de Zacatecas 'Francisco García Salinas', Zacatecas 98160, Mexico.,Bioengineering Laboratory, Unidad Academica de Ingenieria Electrica, Universidad Autonoma de Zacatecas, Zacatecas 98000, Mexico
| | - Yolanda Ortiz-Castro
- Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud, Universidad Autonoma de Zacatecas 'Francisco García Salinas', Zacatecas 98160, Mexico
| | - Panfilo Mauricio-Saucedo
- Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud, Universidad Autonoma de Zacatecas 'Francisco García Salinas', Zacatecas 98160, Mexico.,Departamento de Enseñanza e Investigación, Hospital General Zacatecas 'Luz González Cosío', Servicios de Salud de Zacatecas, Zacatecas 98160, Mexico
| | - Edith Cardenas-Vargas
- Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud, Universidad Autonoma de Zacatecas 'Francisco García Salinas', Zacatecas 98160, Mexico.,Departamento de Enseñanza e Investigación, Hospital General Zacatecas 'Luz González Cosío', Servicios de Salud de Zacatecas, Zacatecas 98160, Mexico
| | - Mariana Diaz-Baez
- Department of Gastroenterology, Hospital General de Zacatecas, Instituto de Seguridad y Servicios Sociales Para Los Trabajadores del Estado, Zacatecas 98000, Mexico
| | - Miguel A Cid-Baez
- Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud, Universidad Autonoma de Zacatecas 'Francisco García Salinas', Zacatecas 98160, Mexico.,Bioengineering Laboratory, Unidad Academica de Ingenieria Electrica, Universidad Autonoma de Zacatecas, Zacatecas 98000, Mexico
| | - Rodrigo Castañeda-Miranda
- Bioengineering Laboratory, Unidad Academica de Ingenieria Electrica, Universidad Autonoma de Zacatecas, Zacatecas 98000, Mexico
| | - Jose Manuel Ortiz-Rodriguez
- Bioengineering Laboratory, Unidad Academica de Ingenieria Electrica, Universidad Autonoma de Zacatecas, Zacatecas 98000, Mexico
| | - Luis Octavio Solis-Sanchez
- Bioengineering Laboratory, Unidad Academica de Ingenieria Electrica, Universidad Autonoma de Zacatecas, Zacatecas 98000, Mexico
| | - Margarita L Martinez-Fierro
- Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud, Universidad Autonoma de Zacatecas 'Francisco García Salinas', Zacatecas 98160, Mexico.,Bioengineering Laboratory, Unidad Academica de Ingenieria Electrica, Universidad Autonoma de Zacatecas, Zacatecas 98000, Mexico
| |
Collapse
|
35
|
Petrus P, Bialesova L, Checa A, Kerr A, Naz S, Bäckdahl J, Gracia A, Toft S, Dahlman-Wright K, Hedén P, Dahlman I, Wheelock CE, Arner P, Mejhert N, Gao H, Rydén M. Adipocyte Expression of SLC19A1 Links DNA Hypermethylation to Adipose Tissue Inflammation and Insulin Resistance. J Clin Endocrinol Metab 2018; 103:710-721. [PMID: 29121255 DOI: 10.1210/jc.2017-01382] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 11/02/2017] [Indexed: 02/07/2023]
Abstract
CONTEXT Insulin resistance (IR) is promoted by a chronic low-grade inflammation in white adipose tissue (WAT). The latter might be regulated through epigenetic mechanisms such as DNA methylation. The one carbon cycle (1CC) is a central metabolic process governing DNA methylation. OBJECTIVE To identify adipocyte-expressed 1CC genes linked to WAT inflammation, IR, and their causal role. DESIGN Cohort study. SETTING Outpatient academic clinic. PARTICIPANTS Obese and nonobese subjects. METHODS Gene expression and DNA methylation arrays were performed in subcutaneous WAT and isolated adipocytes. In in vitro differentiated human adipocytes, gene knockdown was achieved by small interfering RNA, and analyses included microarray, quantitative polymerase chain reaction, DNA methylation by enzyme-linked immunosorbent assay and pyrosequencing, protein secretion by enzyme-linked immunosorbent assay, targeted metabolomics, and luciferase reporter and thermal shift assays. MAIN OUTCOME MEASURES Effects on adipocyte inflammation. RESULTS In adipocytes from obese individuals, global DNA hypermethylation was associated positively with gene expression of proinflammatory pathways. Among the 1CC genes, IR in vivo and proinflammatory gene expression in WAT were most strongly and inversely associated with SLC19A1, a gene encoding a membrane folate carrier. SLC19A1 knockdown in human adipocytes perturbed intracellular 1CC metabolism, induced global DNA hypermethylation, and increased expression of proinflammatory genes. Several CpG loci linked SLC19A1 to inflammation; validation studies were focused on the chemokine C-C motif chemokine ligand 2 (CCL2) in which methylation in the promoter (cg12698626) regulated CCL2 expression and CCL2 secretion through altered transcriptional activity. CONCLUSIONS Reduced SLC19A1 expression in human adipocytes induces DNA hypermethylation, resulting in increased expression of specific proinflammatory genes, including CCL2. This constitutes an epigenetic mechanism that might link dysfunctional adipocytes to WAT inflammation and IR.
Collapse
Affiliation(s)
- Paul Petrus
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lucia Bialesova
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Antonio Checa
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Alastair Kerr
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Shama Naz
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jesper Bäckdahl
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ana Gracia
- Department of Nutrition and Food Science, University of Basque Country (UPV/EHU), Vitoria, Spain
- CIBER Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Sofia Toft
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karin Dahlman-Wright
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Per Hedén
- Department of Plastic Surgery, Akademikliniken, Stockholm, Sweden
| | - Ingrid Dahlman
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Craig E Wheelock
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Peter Arner
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Niklas Mejhert
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hui Gao
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Rydén
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
36
|
Integrated Immunomodulatory Mechanisms through which Long-Chain n-3 Polyunsaturated Fatty Acids Attenuate Obese Adipose Tissue Dysfunction. Nutrients 2017; 9:nu9121289. [PMID: 29186929 PMCID: PMC5748740 DOI: 10.3390/nu9121289] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/14/2017] [Accepted: 11/16/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity is a global health concern with rising prevalence that increases the risk of developing other chronic diseases. A causal link connecting overnutrition, the development of obesity and obesity-associated co-morbidities is visceral adipose tissue (AT) dysfunction, characterized by changes in the cellularity of various immune cell populations, altered production of inflammatory adipokines that sustain a chronic state of low-grade inflammation and, ultimately, dysregulated AT metabolic function. Therefore, dietary intervention strategies aimed to halt the progression of obese AT dysfunction through any of the aforementioned processes represent an important active area of research. In this connection, fish oil-derived dietary long-chain n-3 polyunsaturated fatty acids (PUFA) in the form of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have been demonstrated to attenuate obese AT dysfunction through multiple mechanisms, ultimately affecting AT immune cellularity and function, adipokine production, and metabolic signaling pathways, all of which will be discussed herein.
Collapse
|
37
|
Barreto G, Sandelin J, Salem A, Nordström DC, Waris E. Toll-like receptors and their soluble forms differ in the knee and thumb basal osteoarthritic joints. Acta Orthop 2017; 88:326-333. [PMID: 28093922 PMCID: PMC5434604 DOI: 10.1080/17453674.2017.1281058] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background and purpose - Although the pathogenesis of osteoarthritis (OA) is not well understood, chondrocyte-mediated inflammatory responses (triggered by the activation of innate immune receptors by damage-associated molecules) are thought to be involved. We examined the relationship between Toll-like receptors (TLRs) and OA in cartilage from 2 joints differing in size and mechanical loading: the first carpometacarpal (CMC-I) and the knee. Patients and methods - Samples of human cartilage obtained from OA CMC-I and knee joints were immunostained for TLRs (1-9) and analyzed using histomorphometry and principal component analysis (PCA). mRNA expression levels were analyzed with RT-PCR. Collected synovial fluid (SF) samples were screened for the presence of soluble forms of TLR2 and TLR4 by enzyme-linked immunosorbent assay (ELISA). Results - In contrast to knee OA, TLR expression in CMC-I OA did not show grade-dependent overall profile changes, but PCA revealed that TLR expression profiles clustered according to their cellular compartment organization. Protein levels of TLR4 were substantially higher in knee OA than in CMC-I OA, while the opposite was the case at the mRNA level. ELISA assays confirmed the presence of soluble forms of TLR2 and TLR4 in SF, with sTLR4 being considerably higher in CMC-I OA than in knee OA. Interpretation - We observed that TLRs are differentially expressed in OA cartilage, depending on the joint. Soluble forms of TLR2 and TLR4 were detected for the first time in SF of osteoarthritic joints, with soluble TLR4 being differentially expressed. Together, our results suggest that negative regulatory mechanisms of innate immunity may be involved in the pathomolecular mechanisms of osteoarthritis.
Collapse
Affiliation(s)
- Goncalo Barreto
- Clinicum, Faculty of Medicine, University of Helsinki;,ORTON Orthopaedic Institute of the Invalid Foundation;,Correspondence:
| | | | - Abdelhakim Salem
- Clinicum, Faculty of Medicine, University of Helsinki;,Institute of Dentistry, Clinicum, University of Helsinki
| | - Dan C Nordström
- Department of Rheumatology, Helsinki University and Helsinki University Hospital
| | - Eero Waris
- Department of Hand Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
38
|
Innate Immunity of Adipose Tissue in Rodent Models of Local and Systemic Staphylococcus aureus Infection. Mediators Inflamm 2017; 2017:5315602. [PMID: 28428684 PMCID: PMC5385907 DOI: 10.1155/2017/5315602] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/13/2017] [Indexed: 12/31/2022] Open
Abstract
Background. The role of adipose tissue in systemic inflammation during bacterial infection is unclear. Effects of Staphylococcus aureus infection on adipocytes in rodent models of experimental endocarditis and peritonitis, the impact of S. aureus infection on gene expression in epididymal and subcutaneous adipose tissue, and effects of S. aureus infection on the toll-like receptor-2- (TLR2-) cathelicidin pathway in vivo and in vitro were investigated. Material and methods. The rat model of catheter-induced S. aureus endocarditis and the mouse model of S. aureus-induced peritonitis were used for infection experiments, gene expression profiling in adipose tissue, and measurement of cytokines. 3T3-L1 adipocytes were analyzed for expression of the TLR2-cathelicidin pathway. Results. Upon systemic bacterial infection by S. aureus, there is a shift from anti- to proinflammatory cytokines in serum and in adipose tissue gene expression. The TLR2-cathelicidin pathway is increasingly expressed during adipocyte differentiation in vitro and is induced upon stimulation by synthetic lipopeptides. Conclusions. Systemic infection by Gram-positive bacteria induces proinflammatory transformation of adipose tissue sites distinct from infection sites, documented on the levels of gene expression and secreted mediators. The TLR2-cathelicidine pathway is expressed and highly inducible in adipocytes in vitro. Lipopeptides are important immune-modulators of adipocytes in both gene expression and protein secretion.
Collapse
|
39
|
Boa BCS, Yudkin JS, van Hinsbergh VWM, Bouskela E, Eringa EC. Exercise effects on perivascular adipose tissue: endocrine and paracrine determinants of vascular function. Br J Pharmacol 2017; 174:3466-3481. [PMID: 28147449 DOI: 10.1111/bph.13732] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 01/20/2017] [Accepted: 01/23/2017] [Indexed: 12/11/2022] Open
Abstract
Obesity is a global epidemic, accompanied by increased risk of type 2 diabetes and cardiovascular disease. Adipose tissue hypertrophy is associated with adipose tissue inflammation, which alters the secretion of adipose tissue-derived bioactive products, known as adipokines. Adipokines determine vessel wall properties such as smooth muscle tone and vessel wall inflammation. Exercise is a mainstay of prevention of chronic, non-communicable diseases, type 2 diabetes and cardiovascular disease in particular. Aside from reducing adipose tissue mass, exercise has been shown to reduce inflammatory activity in this tissue. Mechanistically, contracting muscles release bioactive molecules known as myokines, which alter the metabolic phenotype of adipose tissue. In adipose tissue, myokines induce browning, enhance fatty acid oxidation and improve insulin sensitivity. In the past years, the perivascular adipose tissue (PVAT) which surrounds the vasculature, has been shown to control vascular tone and inflammation through local release of adipokines. In obesity, an increase in mass and inflammation of PVAT culminate in dysregulation of adipokine secretion, which contributes to vascular dysfunction. This review describes our current understanding of the mechanisms by which active muscles interact with adipose tissue and improve vascular function. Aside from the exercise-dependent regulation of canonical adipose tissue function, we will focus on the interactions between skeletal muscle and PVAT and the role of novel myokines, such as IL-15, FGF21 and irisin, in these interactions. LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- B C S Boa
- Department of Physiology, VU University Medical Centre, Amsterdam, The Netherlands.,Laboratory for Clinical and Experimental Research on Vascular Biology (BioVasc), Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J S Yudkin
- Department of Medicine, University College London, London, UK
| | - V W M van Hinsbergh
- Department of Physiology, VU University Medical Centre, Amsterdam, The Netherlands
| | - E Bouskela
- Laboratory for Clinical and Experimental Research on Vascular Biology (BioVasc), Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - E C Eringa
- Department of Physiology, VU University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Krautbauer S, Haberl EM, Eisinger K, Pohl R, Rein-Fischboeck L, Rentero C, Alvarez-Guaita A, Enrich C, Grewal T, Buechler C, Neumeier M. Annexin A6 regulates adipocyte lipid storage and adiponectin release. Mol Cell Endocrinol 2017; 439:419-430. [PMID: 27702590 DOI: 10.1016/j.mce.2016.09.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 09/30/2016] [Accepted: 09/30/2016] [Indexed: 12/28/2022]
Abstract
Lipid storage and adipokine secretion are critical features of adipocytes. Annexin A6 (AnxA6) is a lipid-binding protein regulating secretory pathways and its role in adiponectin release was examined. The siRNA-mediated AnxA6 knock-down in 3T3-L1 preadipocytes impaired proliferation, and differentiation of AnxA6-depleted cells to mature adipocytes was associated with higher soluble adiponectin and increased triglyceride storage. The latter was partly attributed to reduced lipolysis. Accordingly, AnxA6 overexpression in 3T3-L1 adipocytes lowered cellular triglycerides and adiponectin secretion. Indeed, serum adiponectin was increased in AnxA6 deficient mice. Expression analysis identified AnxA6 protein to be more abundant in intra-abdominal compared to subcutaneous adipose tissues of mice and men. AnxA6 protein levels increased in white adipose tissues of obese mice and here, levels were highest in subcutaneous fat. AnxA6 protein in adipocytes was upregulated by oxidative stress which might trigger AnxA6 induction in adipose tissues and contribute to impaired fat storage and adiponectin release.
Collapse
Affiliation(s)
- Sabrina Krautbauer
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany
| | - Elisabeth M Haberl
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany
| | - Kristina Eisinger
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany
| | - Rebekka Pohl
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany
| | - Lisa Rein-Fischboeck
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany
| | - Carles Rentero
- Departament de Biologia Cellular, Immunologia i Neurociències, Facultat de Medicina, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
| | - Anna Alvarez-Guaita
- Departament de Biologia Cellular, Immunologia i Neurociències, Facultat de Medicina, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
| | - Carlos Enrich
- Departament de Biologia Cellular, Immunologia i Neurociències, Facultat de Medicina, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
| | - Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, 2006, Australia
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany.
| | - Markus Neumeier
- Department of Internal Medicine I, Regensburg University Hospital, 93042 Regensburg, Germany
| |
Collapse
|
41
|
Rozati R, Allauddin N. Single Nucleotide Polymorphisms in CD14 and Toll-like Receptor 4 Genes in Patients with Polycystic Ovarian Syndrome. ACTA ACUST UNITED AC 2017. [DOI: 10.5005/jp-journals-10006-1518] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
ABSTRACT
Introduction
Polycystic ovary syndrome (PCOS) is a complex disease having genetic, immunologic, and environmental components, and candidate genes on innate immunity have been hypothesized to be involved in its etiology. We examined the possible association of CD14 and toll-like receptor 4 (TLR4) polymorphisms with PCOS.
Materials and methods
A total of 219 women with PCOS and 272 healthy women were recruited in the study. Their samples were genotyped for the polymorphism of CD14 and TLR4 genes.
Results
The distributions of genotypes of both polymorphisms were found to be significant in women with PCOS compared with controls. The distributions of alleles were also found to be predominant in PCOS compared with controls.
Conclusion
Polymorphisms in CD14-159C>T and TLR4-299A>G significantly increased susceptibility to PCOS. Further studies with larger sample sizes are warranted to confirm these findings.
How to cite this article
Allauddin N, Rozati R. Single Nucleotide Polymorphisms in CD14 and Toll-like Receptor 4 Genes in Patients with Polycystic Ovarian Syndrome. J South Asian Feder Obst Gynae 2017;9(4):304-307.
Collapse
|
42
|
Survival and Inflammatory Response in Adipose-derived Mesenchymal Stem Cell-enriched Mouse Fat Grafts. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2016; 4:e1110. [PMID: 28293494 PMCID: PMC5222639 DOI: 10.1097/gox.0000000000001110] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/12/2016] [Indexed: 01/22/2023]
Abstract
Supplemental Digital Content is available in the text. Background: Adipose tissue-derived mesenchymal stem cells (ATMSCs) are currently used in grafting procedures in a number of clinical trials. The reconstructive role of such cells in fat graft enrichment is largely unclear. This study was undertaken to assess survival and inflammatory response in fat grafts enriched with ATMSCs in mice. Methods: ATMSC-enriched adipose tissue was grafted subcutaneously in a clinically relevant manner in mice, and survival and inflammatory response were determined by bioluminescence imaging of transgenic tissue constitutively expressing luciferase or driven by inflammation in wild-type animals. Results: Only a minor fraction of ATMSCs transplanted subcutaneously were found to survive long term, yet fat grafts enriched with ATMSCs showed improved survival for a limited period, compared with no enrichment. NF-κB activity was transiently increased in ATMSC-enriched grafts, and the grafts responded adequately to a proinflammatory stimulus. In one animal, cells originating from the subcutaneous graft were found at a site of inflammation distant from the site of engraftment. Conclusion: ATMSCs display limited subcutaneous survival. Still, ATMSC enrichment may improve the outcome of adipose tissue grafting procedures by facilitating short-term graft survival and adequate inflammatory responses. Migration of cells from grafted adipose tissue requires further investigation.
Collapse
|
43
|
Mattos RT, Medeiros NI, Menezes CA, Fares RCG, Franco EP, Dutra WO, Rios-Santos F, Correa-Oliveira R, Gomes JAS. Chronic Low-Grade Inflammation in Childhood Obesity Is Associated with Decreased IL-10 Expression by Monocyte Subsets. PLoS One 2016; 11:e0168610. [PMID: 27977792 PMCID: PMC5158089 DOI: 10.1371/journal.pone.0168610] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 12/03/2016] [Indexed: 12/19/2022] Open
Abstract
Chronic low-grade inflammation is related to the development of comorbidities and poor prognosis in obesity. Monocytes are main sources of cytokines and play a pivotal role in inflammation. We evaluated monocyte frequency, phenotype and cytokine profile of monocyte subsets, to determine their association with the pathogenesis of childhood obesity. Children with obesity were evaluated for biochemical and anthropometric parameters. Monocyte subsets were characterized by flow cytometry, considering cytokine production and activation/recognition molecules. Correlation analysis between clinical parameters and immunological data delineated the monocytes contribution for low-grade inflammation. We observed a higher frequency of non-classical monocytes in the childhood obesity group (CO) than normal-weight group (NW). All subsets displayed higher TLR4 expression in CO, but their recognition and antigen presentation functions seem to be diminished due to lower expression of CD40, CD80/86 and HLA-DR. All subsets showed a lower expression of IL-10 in CO and correlation analyses showed changes in IL-10 expression profile. The lower expression of IL-10 may be decisive for the maintenance of the low-grade inflammation status in CO, especially for alterations in non-classical monocytes profile. These cells may contribute to supporting inflammation and loss of regulation in the immune response of children with obesity.
Collapse
Affiliation(s)
- Rafael T. Mattos
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Nayara I. Medeiros
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
- Laboratório de Imunologia Celular e Molecular, Centro de Pesquisa René Rachou, FIOCRUZ, Belo Horizonte, MG, Brasil
| | - Carlos A. Menezes
- Departamento de Genética, Universidade Estadual de Santa Cruz, Ilhéus, BA, Brasil
- Serviço de Medicina Preventiva da Unimed, Aracaju, SE, Brasil
| | - Rafaelle C. G. Fares
- Laboratório de Imunologia Celular e Molecular, Centro de Pesquisa René Rachou, FIOCRUZ, Belo Horizonte, MG, Brasil
| | - Eliza P. Franco
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Walderez O. Dutra
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
- Instituto Nacional de Ciência e Tecnologia em Doenças Topicais—INCT-DT
| | - Fabrício Rios-Santos
- Faculdade de Medicina, Departamento de Ciências Básicas da Saúde, Universidade Federal de Mato Grasso, Cuiabá, MT, Brasil
| | - Rodrigo Correa-Oliveira
- Laboratório de Imunologia Celular e Molecular, Centro de Pesquisa René Rachou, FIOCRUZ, Belo Horizonte, MG, Brasil
- Instituto Nacional de Ciência e Tecnologia em Doenças Topicais—INCT-DT
| | - Juliana A. S. Gomes
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
- * E-mail:
| |
Collapse
|
44
|
Vlodavsky I, Singh P, Boyango I, Gutter-Kapon L, Elkin M, Sanderson RD, Ilan N. Heparanase: From basic research to therapeutic applications in cancer and inflammation. Drug Resist Updat 2016; 29:54-75. [PMID: 27912844 DOI: 10.1016/j.drup.2016.10.001] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Heparanase, the sole heparan sulfate degrading endoglycosidase, regulates multiple biological activities that enhance tumor growth, angiogenesis and metastasis. Heparanase expression is enhanced in almost all cancers examined including various carcinomas, sarcomas and hematological malignancies. Numerous clinical association studies have consistently demonstrated that upregulation of heparanase expression correlates with increased tumor size, tumor angiogenesis, enhanced metastasis and poor prognosis. In contrast, knockdown of heparanase or treatments of tumor-bearing mice with heparanase-inhibiting compounds, markedly attenuate tumor progression further underscoring the potential of anti-heparanase therapy for multiple types of cancer. Heparanase neutralizing monoclonal antibodies block myeloma and lymphoma tumor growth and dissemination; this is attributable to a combined effect on the tumor cells and/or cells of the tumor microenvironment. In fact, much of the impact of heparanase on tumor progression is related to its function in mediating tumor-host crosstalk, priming the tumor microenvironment to better support tumor growth, metastasis and chemoresistance. The repertoire of the physio-pathological activities of heparanase is expanding. Specifically, heparanase regulates gene expression, activates cells of the innate immune system, promotes the formation of exosomes and autophagosomes, and stimulates signal transduction pathways via enzymatic and non-enzymatic activities. These effects dynamically impact multiple regulatory pathways that together drive inflammatory responses, tumor survival, growth, dissemination and drug resistance; but in the same time, may fulfill some normal functions associated, for example, with vesicular traffic, lysosomal-based secretion, stress response, and heparan sulfate turnover. Heparanase is upregulated in response to chemotherapy in cancer patients and the surviving cells acquire chemoresistance, attributed, at least in part, to autophagy. Consequently, heparanase inhibitors used in tandem with chemotherapeutic drugs overcome initial chemoresistance, providing a strong rationale for applying anti-heparanase therapy in combination with conventional anti-cancer drugs. Heparin-like compounds that inhibit heparanase activity are being evaluated in clinical trials for various types of cancer. Heparanase neutralizing monoclonal antibodies are being evaluated in pre-clinical studies, and heparanase-inhibiting small molecules are being developed based on the recently resolved crystal structure of the heparanase protein. Collectively, the emerging premise is that heparanase expressed by tumor cells, innate immune cells, activated endothelial cells as well as other cells of the tumor microenvironment is a master regulator of the aggressive phenotype of cancer, an important contributor to the poor outcome of cancer patients and a prime target for therapy.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel.
| | - Preeti Singh
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Ilanit Boyango
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Lilach Gutter-Kapon
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Michael Elkin
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ralph D Sanderson
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Neta Ilan
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| |
Collapse
|
45
|
Karrasch T, Schaeffler A. Adipokines and the role of visceral adipose tissue in inflammatory bowel disease. Ann Gastroenterol 2016; 29:424-438. [PMID: 27708507 PMCID: PMC5049548 DOI: 10.20524/aog.2016.0077] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/22/2016] [Indexed: 12/20/2022] Open
Abstract
Recently, adipocytes have been recognized as actively participating in local and systemic immune responses via the secretion of peptides detectable in relevant levels in the systemic circulation, the so-called "adipo(cyto)kines". Multiple studies appearing within the last 10-15 years have focused on the possible impact of adipose tissue depots on inflammatory bowel disease (IBD). Consequently, various hypotheses regarding the role of different adipokines in inflammatory diseases in general and in intestinal inflammatory processes in particular have been developed and have been further refined in recent years. After a focused summary of the data reported concerning the impact of visceral adipose tissue on IBD, such as Crohn's disease and ulcerative colitis, our review focuses on recent developments indicating that adipocytes as part of the innate immune system actively participate in antimicrobial host defenses in the context of intestinal bacterial translocation, which are of utmost importance for the homeostasis of the whole organism. Modulators of adipose tissue function and regulators of adipokine secretion, as well as modifiers of adipocytic pattern recognition molecules, might represent future potential drug targets in IBD.
Collapse
Affiliation(s)
- Thomas Karrasch
- Department of Internal Medicine III, Giessen University Hospital, Germany
| | - Andreas Schaeffler
- Department of Internal Medicine III, Giessen University Hospital, Germany
| |
Collapse
|
46
|
Interaction between gut microbiota and toll-like receptor: from immunity to metabolism. J Mol Med (Berl) 2016; 95:13-20. [PMID: 27639584 PMCID: PMC5225216 DOI: 10.1007/s00109-016-1474-4] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/15/2016] [Accepted: 09/08/2016] [Indexed: 02/06/2023]
Abstract
The human gut contains trillions of commensal bacteria, and similar to pathogenic bacteria, the gut microbes and their products can be recognized by toll-like receptors (TLRs). It is well acknowledged that the interaction between gut microbiota and the local TLRs help to maintain the homeostasis of intestinal immunity. High-fat intake or obesity can weaken gut integrity leading to the penetration of gut microbiota or their bacterial products into the circulation, leading to the activation of TLRs on immune cells and subsequently low-grade systemic inflammation in host. Metabolic cells including hepatocytes and adipocytes also express TLRs. Although they are able to produce and secrete inflammatory molecules, the effectiveness remains low compared with the immune cells embedded in the liver and adipose tissue. The interaction of TLRs in these metabolic cells or organs with gut microbiota remains unclear, but a few studies have suggested that the functions of these TLRs are related to metabolism. Alteration of the gut microbiota is associated with body weight change and adiposity in human, and the interaction between the commensal gut microbiota and TLRs may possibly involve both metabolic and immunological regulation. In this review, we will summarize the current findings on the relationship between TLRs and gut microbiota with a focus on metabolic regulation and discuss how such interaction participates in host metabolism.
Collapse
|
47
|
Shimizu T, Yamakuchi M, Biswas KK, Aryal B, Yamada S, Hashiguchi T, Maruyama I. HMGB1 is secreted by 3T3-L1 adipocytes through JNK signaling and the secretion is partially inhibited by adiponectin. Obesity (Silver Spring) 2016; 24:1913-21. [PMID: 27430164 DOI: 10.1002/oby.21549] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/13/2016] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Obesity is a chronic inflammatory disease, and adipocytes contribute to obesity-associated inflammation by releasing inflammatory mediators. High mobility group box 1 (HMGB1), a highly conserved DNA-binding protein, mainly localized to cell nuclei, has been recently recognized as an innate pro-inflammatory mediator when released extracellularly. It was hypothesized that HMGB1 is an adipocytokine that acts as an innate pro-inflammatory mediator in white adipose tissue (WAT) of patients with obesity and is associated with insulin resistance. Additionally, it was hypothesized that HMGB1 secretion is regulated by adiponectin. METHODS 3T3-L1 cells were differentiated into mature adipocytes. After tumor necrosis factor-α (TNF-α) stimulation, HMGB1 in culture media was measured. Localizations of HMGB1 in 3T3-L1 adipocytes and human WAT were examined by immunostaining. RESULTS HMGB1 was secreted from TNF-α-induced 3T3-L1 adipocytes through JNK signaling. HMGB1-activated MAP kinases (ERK1/2, JNK) and suppressed insulin-stimulated Akt phosphorylation in 3T3-L1 adipocytes. The cytoplasm in 3T3-L1 adipocytes and adipocytes of WAT from a patient with obesity was intensely stained with HMGB1. Adiponectin partially inhibited TNF-α-induced HMGB1 secretion from 3T3-L1 adipocytes. CONCLUSIONS These findings suggest that HMGB1 is a pro-inflammatory adipocytokine involved in WAT inflammation and insulin resistance in patients with obesity, which may contribute to the progression of metabolic syndrome, and that adiponectin protects against HMGB1-induced adipose tissue inflammation.
Collapse
Affiliation(s)
- Toshiaki Shimizu
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Munekazu Yamakuchi
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kamal Krishna Biswas
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Bibek Aryal
- Cardiovascular and Gastroenterological Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | | | - Teruto Hashiguchi
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Ikuro Maruyama
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
48
|
Shimabukuro M, Sato H, Izaki H, Fukuda D, Uematsu E, Hirata Y, Yagi S, Soeki T, Sakaue H, Kanayama HO, Masuzaki H, Sata M. Depot- and gender-specific expression of NLRP3 inflammasome and toll-like receptors in adipose tissue of cancer patients. Biofactors 2016; 42:397-406. [PMID: 27086574 DOI: 10.1002/biof.1287] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 03/09/2016] [Accepted: 03/10/2016] [Indexed: 02/05/2023]
Abstract
Gender difference in obesity-associated cardiovascular complication could be derived from divergent chronic inflammation. We evaluated depot- and gender-specific regulation of the innate immune system in human adipose tissues. Pair samples were obtained from subcutaneous (SAT) and visceral adipose tissue (VAT) during elective surgery (Male: 35; Female: 27). Expressions of pro- and anti-inflammatory adipocytokines were evaluated by semi-quantitative qPCR. Adipose cell-size distribution was obtained from tissue samples fixed in osmium tetroxide and analyzed by Beckman Coulter Multisizer. Levels of adiponectin were higher in SAT and VAT of female than those of male (P < 0.001 and P = 0.011, respectively). NLRP3, IL1β-IL18, TLR2 were comparable in SAT and VAT between genders. However, TLR4 and TLR9 were increased in female SAT and VAT and HMGB1 in female VAT. Levels of adiponectin were not correlated with mean diameter of adipocyte (φ, μm) in SAT and VAT of male, but negatively well correlated in those of female (r = -0.392 and r = -0.616). Such negative correlations were also observed between levels of TLR2, TLR4, and HMGB1 and φ in female. Levels of NLRP3 and IL1β were positively correlated with φ in male, but not in female. In conclusion, Innate signals were differentially expressed in male and female adipose tissues, suggesting that the depot- and gender-specific signals could be related to gender difference in chronic inflammation. © 2016 BioFactors, 42(4):397-406, 2016.
Collapse
Affiliation(s)
- Michio Shimabukuro
- Department of Cardio-Diabetes Medicine, Institute of Biomedical Sciences, University of Tokushima Graduate School, Tokushima, Japan
- Department of Cardiovascular Medicine, Institute of Biomedical Sciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Hiromi Sato
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Hirofumi Izaki
- Department of Urology, Tokushima Prefectural Central Hospital
| | - Daiju Fukuda
- Department of Cardiovascular Medicine, Institute of Biomedical Sciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Etsuko Uematsu
- Department of Cardiovascular Medicine, Institute of Biomedical Sciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Yoichiro Hirata
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Japan
| | - Shusuke Yagi
- Department of Cardiovascular Medicine, Institute of Biomedical Sciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Takeshi Soeki
- Department of Cardiovascular Medicine, Institute of Biomedical Sciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Hiro-Omi Kanayama
- Department of Urology, Institute of Biomedical Sciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Hiroaki Masuzaki
- Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Institute of Biomedical Sciences, University of Tokushima Graduate School, Tokushima, Japan
| |
Collapse
|
49
|
Tolusso B, Alivernini S, Gigante MR, Ferraccioli G, Gremese E. Biomolecular features of inflammation in obese rheumatoid arthritis patients: management considerations. Expert Rev Clin Immunol 2016; 12:751-62. [PMID: 26950427 DOI: 10.1586/1744666x.2016.1159132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Adipose tissue is an active organ playing a role not only in metabolism but also in immune and inflammatory processes, releasing several pro-inflammatory mediators. This can explain the possible association between obesity and rheumatoid arthritis (RA) and its role in the progression of the disease. Adipose and synovial tissues share common histological features of local inflammation in terms of activation of target tissues infiltrating cells (i.e. myeloid cells). Among the so-called adipocytokines, PEDF and Chemerin orchestrate the cellular cross-talk between adipose and myeloid cells, being possible biomarkers to monitor the effect of weight loss or the decrease of adipose tissue in patients with RA. Moreover, dietary intervention has been demonstrated to reduce Chemerin as well as IL-6 and MCP-1 expression. Finally, epigenetic regulators such as micro-RNAs (i.e. miR-155) are key regulators of myeloid cells activation in RA and obesity as well as in adipocytes. In this review, we will summarize the biological link between obesity/overweight state and RA focusing on pathophysiological mechanisms, consequences and management considerations.
Collapse
Affiliation(s)
- Barbara Tolusso
- a Division of Rheumatology, Institute of Rheumatology , Catholic University of the Sacred Heart , Rome , Italy
| | - Stefano Alivernini
- a Division of Rheumatology, Institute of Rheumatology , Catholic University of the Sacred Heart , Rome , Italy
| | - Maria Rita Gigante
- a Division of Rheumatology, Institute of Rheumatology , Catholic University of the Sacred Heart , Rome , Italy
| | - Gianfranco Ferraccioli
- a Division of Rheumatology, Institute of Rheumatology , Catholic University of the Sacred Heart , Rome , Italy
| | - Elisa Gremese
- a Division of Rheumatology, Institute of Rheumatology , Catholic University of the Sacred Heart , Rome , Italy
| |
Collapse
|
50
|
Durandt C, van Vollenstee FA, Dessels C, Kallmeyer K, de Villiers D, Murdoch C, Potgieter M, Pepper MS. Novel flow cytometric approach for the detection of adipocyte subpopulations during adipogenesis. J Lipid Res 2016; 57:729-42. [PMID: 26830859 PMCID: PMC4808761 DOI: 10.1194/jlr.d065664] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Indexed: 12/17/2022] Open
Abstract
The ability of mesenchymal stromal cells (MSCs) to differentiate into adipocytes provides a cellular model of human origin to study adipogenesis in vitro. One of the major challenges in studying adipogenesis is the lack of tools to identify and monitor the differentiation of various subpopulations within the heterogeneous pool of MSCs. Cluster of differentiation (CD)36 plays an important role in the formation of intracellular lipid droplets, a key characteristic of adipocyte differentiation/maturation. The objective of this study was to develop a reproducible quantitative method to study adipocyte differentiation by comparing two lipophilic dyes [Nile Red (NR) and Bodipy 493/503] in combination with CD36 surface marker staining. We identified a subpopulation of adipose-derived stromal cells that express CD36 at intermediate/high levels and show that combining CD36 cell surface staining with neutral lipid-specific staining allows us to monitor differentiation of adipose-derived stromal cells that express CD36intermediate/high during adipocyte differentiation in vitro. The gradual increase of CD36intermediate/high/NRpositive cells during the 21 day adipogenesis induction period correlated with upregulation of adipogenesis-associated gene expression.
Collapse
Affiliation(s)
- Chrisna Durandt
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Fiona A van Vollenstee
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Carla Dessels
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Karlien Kallmeyer
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Danielle de Villiers
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Candice Murdoch
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Marnie Potgieter
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Michael S Pepper
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|