201
|
Kim ME, Lee JS, Kim TW, Park MH, Kim DH. FoxO6-Mediated TXNIP Induces Lipid Accumulation in the Liver through NLRP3 Inflammasome Activation. Endocrinol Metab (Seoul) 2024; 39:127-139. [PMID: 38417829 PMCID: PMC10901662 DOI: 10.3803/enm.2023.1826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/02/2023] [Accepted: 11/13/2023] [Indexed: 03/01/2024] Open
Abstract
BACKGRUOUND Hepatic steatosis, which involves the excessive accumulation of lipid droplets in hepatocytes, presents a significant global health concern due to its association with obesity and metabolic disorders. Inflammation plays a crucial role in the progression of hepatic steatosis; however, the precise molecular mechanisms responsible for this process remain unknown. METHODS This study investigated the involvement of the nucleotide-binding oligomerization domain-like receptor pyrin domain-containing-3 (NLRP3) inflammasome and the forkhead box O6 (FoxO6) transcription factor in the pathogenesis of hepatic steatosis. We monitored the NLRP3 inflammasome and lipogenesis in mice overexpressing the constitutively active (CA)-FoxO6 allele and FoxO6-null mice. In an in vitro study, we administered palmitate to liver cells overexpressing CA-FoxO6 and measured changes in lipid metabolism. RESULTS We administered palmitate treatment to clarify the mechanisms through which FoxO6 activates cytokine interleukin (IL)-1β through the NLRP3 inflammasome. The initial experiments revealed that dephosphorylation led to palmitate-induced FoxO6 transcriptional activity. Further palmitate experiments showed increased expression of IL-1β and the hepatic NLRP3 inflammasome complex, including adaptor protein apoptotic speck protein containing a caspase recruitment domain (ASC) and pro-caspase-1. Furthermore, thioredoxin-interacting protein (TXNIP), a key regulator of cellular redox conditions upstream of the NLRP3 inflammasome, was induced by FoxO6 in the liver and HepG2 cells. CONCLUSION The findings of this study shed light on the molecular mechanisms underpinning the FoxO6-NLRP3 inflammasome axis in promoting inflammation and lipid accumulation in the liver.
Collapse
Affiliation(s)
- Mi Eun Kim
- Department of Life Sciences, Chosun University College of Natural Science, Gwangju, Korea
| | - Jun Sik Lee
- Department of Life Sciences, Chosun University College of Natural Science, Gwangju, Korea
| | - Tae Won Kim
- Department of Pharmacy, Kyungsung University College of Pharmacy, Busan, Korea
| | - Min Hi Park
- Department of Pharmacy, Kyungsung University College of Pharmacy, Busan, Korea
| | - Dae Hyun Kim
- Department of Food Science & Technology, Pusan National University College of Natural Resources and Life Science, Miryang, Korea
| |
Collapse
|
202
|
Krajewski PK, Tsoukas M, Szepietowski JC. Pathological and Therapeutical Implications of Pyroptosis in Psoriasis and Hidradenitis Suppurativa: A Narrative Review. Curr Issues Mol Biol 2024; 46:663-676. [PMID: 38248345 PMCID: PMC10814322 DOI: 10.3390/cimb46010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
This manuscript explores the role of pyroptosis, an inflammatory programmed cell death, in the pathogenesis of two chronic dermatoses, psoriasis and hidradenitis suppurativa (HS). The diseases, though clinically diverse, share common pathogenetic pathways involving the unbalanced interaction between the adaptive and innate immune systems. This review focuses on the molecular changes in psoriatic and HS skin, emphasizing the activation of dendritic cells, secretion of interleukins (IL-17, IL-22, and TNF-α), and the involvement of inflammasomes, particularly NLRP3. This manuscript discusses the role of caspases, especially caspase-1, in driving pyroptosis and highlights the family of gasdermins (GSDMs) as key players in the formation of pores leading to cell rupture and the release of proinflammatory signals. This study delves into the potential therapeutic implications of targeting pyroptosis in psoriasis and HS, examining existing medications like biologics and Janus kinase inhibitors. It also reviews the current limitations and challenges in developing therapies that selectively target pyroptosis. Additionally, the manuscript explores the role of pyroptosis in various inflammatory disorders associated with psoriasis and HS, such as inflammatory bowel disease, diabetes mellitus, and cardiovascular disorders. The review concludes by emphasizing the need for further research to fully elucidate the pathomechanisms of these dermatoses and develop effective, targeted therapies.
Collapse
Affiliation(s)
- Piotr K. Krajewski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Chalubinskiego 1, 50-368 Wroclaw, Poland;
| | - Maria Tsoukas
- Department of Dermatology, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Jacek C. Szepietowski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Chalubinskiego 1, 50-368 Wroclaw, Poland;
| |
Collapse
|
203
|
Panyod S, Wu WK, Hu MY, Huang HS, Chen RA, Chen YH, Shen TCD, Ho CT, Liu CJ, Chuang HL, Huang CC, Wu MS, Sheen LY. Healthy diet intervention reverses the progression of NASH through gut microbiota modulation. Microbiol Spectr 2024; 12:e0186823. [PMID: 38018983 PMCID: PMC10782987 DOI: 10.1128/spectrum.01868-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/27/2023] [Indexed: 11/30/2023] Open
Abstract
IMPORTANCE The link between gut microbiota and diet is crucial in the development of non-alcoholic steatohepatitis (NASH). This study underscores the essential role of a healthy diet in preventing and treating NASH by reversing obesity, lipidemia, and gut microbiota dysbiosis. Moreover, the supplementation of functional food or drug to the diet can provide additional advantages by inhibiting hepatic inflammation through the modulation of the hepatic inflammasome signaling pathway and partially mediating the gut microbiota and lipopolysaccharide signaling pathway. This study highlights the importance of adopting healthy dietary habits in treating NASH and proposes that supplementing with ginger essential oil or obeticholic acid may offer additional benefits. Nonetheless, further clinical studies are necessary to validate these findings.
Collapse
Affiliation(s)
- Suraphan Panyod
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
- Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan
| | - Wei-Kai Wu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Meng-Yun Hu
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Huai-Syuan Huang
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Rou-An Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yi-Hsun Chen
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ting-Chin David Shen
- Division of Gastroenterology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey, USA
| | - Chun-Jen Liu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Chi-Chang Huang
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan City, Taiwan
| | - Ming-Shiang Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Lee-Yan Sheen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
- Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan
- National Taiwan University, National Center for Food Safety Education and Research, Taipei, Taiwan
| |
Collapse
|
204
|
Zhao Y, Zhao H, Li L, Yu S, Liu M, Jiang L. Ceramide on the road to insulin resistance and immunometabolic disorders in transition dairy cows: driver or passenger? Front Immunol 2024; 14:1321597. [PMID: 38274826 PMCID: PMC10808295 DOI: 10.3389/fimmu.2023.1321597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Dairy cows must undergo profound metabolic and endocrine adaptations during their transition period to meet the nutrient requirements of the developing fetus, parturition, and the onset of lactation. Insulin resistance in extrahepatic tissues is a critical component of homeorhetic adaptations in periparturient dairy cows. However, due to increased energy demands at calving that are not followed by a concomitant increase in dry matter intake, body stores are mobilized, and the risk of metabolic disorders dramatically increases. Sphingolipid ceramides involved in multiple vital biological processes, such as proliferation, differentiation, apoptosis, and inflammation. Three typical pathways generate ceramide, and many factors contribute to its production as part of the cell's stress response. Based on lipidomic profiling, there has generally been an association between increased ceramide content and various disease outcomes in rodents. Emerging evidence shows that ceramides might play crucial roles in the adaptive metabolic alterations accompanying the initiation of lactation in dairy cows. A series of studies also revealed a negative association between circulating ceramides and systemic insulin sensitivity in dairy cows experiencing severe negative energy balance. Whether ceramide acts as a driver or passenger in the metabolic stress of periparturient dairy cows is an unknown but exciting topic. In the present review, we discuss the potential roles of ceramides in various metabolic dysfunctions and the impacts of their perturbations. We also discuss how this novel class of bioactive sphingolipids has drawn interest in extrahepatic tissue insulin resistance and immunometabolic disorders in transition dairy cows. We also discuss the possible use of ceramide as a new biomarker for predicting metabolic diseases in cows and highlight the remaining problems.
Collapse
Affiliation(s)
| | | | | | | | | | - Linshu Jiang
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| |
Collapse
|
205
|
Johnson DC, Bryan EE, Burris ES, Dilger RN, Harsh BN, Dilger AC. Effects of maternal inflammation on growth performance, carcass characteristics, and meat quality of offspring pigs. J Anim Sci 2024; 102:skae215. [PMID: 39066604 PMCID: PMC11336995 DOI: 10.1093/jas/skae215] [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: 05/13/2024] [Accepted: 07/25/2024] [Indexed: 07/28/2024] Open
Abstract
The objective of this research was to determine the effects of mid-gestational maternal inflammation on performance, carcass characteristics, and meat quality of offspring. Pregnant gilts were administered either lipopolysaccharide (LPS; n = 7) or saline (CON, n = 7) from days 70 to 84 of gestation. Gilts assigned to the LPS treatment were administered an intravenous injection of reconstituted LPS every other day with a beginning dose of 10 μg LPS/kg body weight and subsequent doses increasing by 12%, while CON gilts received intravenous injections of comparable volumes of saline. Gilts farrowed naturally, and at day 66 of age, a total of 59 pigs, both barrows and gilts began a 3-phase feeding regimen designed to meet or exceed nutrient requirements for growing-finishing pigs. Pigs were weighed on days 0, 35, 70, and 105 of the finishing trial to determine average daily gain, average daily feed intake, and gain-to-feed ratio (G:F). On day 106, pigs were slaughtered under the supervision of the U.S. Department of Agriculture Food Safety Inspection Service. Ending live weight, hot carcass weight, and dressing percentage were determined. The left side of carcasses was weighed and fabricated to determine carcass cutting yields. The semitendinosus was collected for histological samples. Fresh belly characteristics and loin quality were measured. Two chops were collected for Warner-Bratzler shear force and proximate analysis. No differences (P ≥ 0.13) between LPS and CON pigs were observed for growth performance in phases 1, 2, 3, or overall (days 0 to 105) performance with the exception of overall G:F reduced in CON pigs compared with LPS pigs (P = 0.03). There was a tendency for carcass yield to be reduced (P = 0.06; 0.82% units) in LPS pigs compared with CON pigs. Additionally, longissimus muscle area tended to be reduced (P = 0.10) 2.27 cm2 in LPS compared with CON pigs. Loin chop quality traits including instrumental color, subjective color, marbling, firmness, pH, and drip loss were not different (P ≥ 0.09) between LPS and CON pigs. Fresh belly characteristics were not different (P ≥ 0.22) between LPS and CON pigs. There were no differences in primal and subprimal weights, except that LPS pigs tended to have a reduction (P ≥ 0.07) in tenderloin and Canadian back weights compared with CON pigs. Furthermore, LPS pigs had no differences (P ≥ 0.25) in muscle fiber composition or size; however, LPS pigs tended (P = 0.10) to have a 13% reduction in estimated muscle fibers number compared with CON pigs. In summary, mid-gestational inflammation did not result in reduced meat quality, growth performance, or carcass yields of offspring.
Collapse
Affiliation(s)
- Danielle C Johnson
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Erin E Bryan
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Elli S Burris
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Ryan N Dilger
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Bailey N Harsh
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Anna C Dilger
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| |
Collapse
|
206
|
Dicks LMT. How important are fatty acids in human health and can they be used in treating diseases? Gut Microbes 2024; 16:2420765. [PMID: 39462280 PMCID: PMC11520540 DOI: 10.1080/19490976.2024.2420765] [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: 08/07/2024] [Revised: 09/19/2024] [Accepted: 10/19/2024] [Indexed: 10/29/2024] Open
Abstract
Most of the short-chain fatty acids (SCFAs) are produced by Bifidobacterium, Lactobacillus, Lachnospiraceae, Blautia, Coprococcus, Roseburia, Facealibacterium and Oscillospira. Butyrate (C4H7O2-) supplies 70% of energy to intestinal epithelial cells (IECs), supports tight-junction protein formation, induces the production of inflammatory cytokines, and inhibits histone deacetylase (HDAC). Butyrate is also associated with the recovery of brain trauma, improvement of dementia, the alleviation of autoimmune encephalitis, and several intestinal disorders. Low levels of SCFAs are associated with hypertension, cardiovascular disease (CVD), strokes, obesity, and diabetes mellitus. Cis-palmitoleic acid (C16H30O2), a mono-unsaturated fatty acid (MUFA), increases insulin sensitivity and reduces the risk of developing CVD. Lipokine palmitoleic acid reduces the expression of pro-inflammatory cytokines IL-1β (pro-IL1β), tumor necrosis factor α (TNF-α), and isoleucine 6 (IL-6). Polyunsaturated fatty acids (PUFAs), such as omega-3 and omega-6, are supplied through the diet. The conversion of PUFAs by cyclooxygenases (COX) and lipoxygenases (LOX) leads to the production of anti-inflammatory prostaglandins and leukotrienes. Oxidation of linoleic acid (LA, C18H32O2), an omega-6 essential fatty acid, leads to the formation of 13-hydroperoxy octadecadienoic acid (13-HPODE, C18H32O4), which induces pro-inflammatory cytokines. Omega-3 PUFAs, such as eicosapentaenoic acid (EPA, C20H30O2) and docosahexaenoic acid (DHA, C22H32O2), lower triglyceride levels, lower the risk of developing some sort of cancers, Alzheimer's disease and dementia. In this review, the importance of SCFAs, MUFAs, PUFAs, and saturated fatty acids (SFAs) on human health is discussed. The use of fatty acids in the treatment of diseases is investigated.
Collapse
Affiliation(s)
- Leon M. T. Dicks
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
207
|
Engin A. Endothelial Dysfunction in Obesity and Therapeutic Targets. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:489-538. [PMID: 39287863 DOI: 10.1007/978-3-031-63657-8_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Parallel to the increasing prevalence of obesity in the world, the mortality from cardiovascular disease has also increased. Low-grade chronic inflammation in obesity disrupts vascular homeostasis, and the dysregulation of adipocyte-derived endocrine and paracrine effects contributes to endothelial dysfunction. Besides the adipose tissue inflammation, decreased nitric oxide (NO)-bioavailability, insulin resistance (IR), and oxidized low-density lipoproteins (oxLDLs) are the main factors contributing to endothelial dysfunction in obesity and the development of cardiorenal metabolic syndrome. While normal healthy perivascular adipose tissue (PVAT) ensures the dilation of blood vessels, obesity-associated PVAT leads to a change in the profile of the released adipo-cytokines, resulting in a decreased vasorelaxing effect. Higher stiffness parameter β, increased oxidative stress, upregulation of pro-inflammatory cytokines, and nicotinamide adenine dinucleotide phosphate (NADP) oxidase in PVAT turn the macrophages into pro-atherogenic phenotypes by oxLDL-induced adipocyte-derived exosome-macrophage crosstalk and contribute to the endothelial dysfunction. In clinical practice, carotid ultrasound, higher leptin levels correlate with irisin over-secretion by human visceral and subcutaneous adipose tissues, and remnant cholesterol (RC) levels predict atherosclerotic disease in obesity. As a novel therapeutic strategy for cardiovascular protection, liraglutide improves vascular dysfunction by modulating a cyclic adenosine monophosphate (cAMP)-independent protein kinase A (PKA)-AMP-activated protein kinase (AMPK) pathway in PVAT in obese individuals. Because the renin-angiotensin-aldosterone system (RAAS) activity, hyperinsulinemia, and the resultant IR play key roles in the progression of cardiovascular disease in obesity, RAAS-targeted therapies contribute to improving endothelial dysfunction. By contrast, arginase reciprocally inhibits NO formation and promotes oxidative stress. Thus, targeting arginase activity as a key mediator in endothelial dysfunction has therapeutic potential in obesity-related vascular comorbidities. Obesity-related endothelial dysfunction plays a pivotal role in the progression of type 2 diabetes (T2D). The peroxisome proliferator-activated receptor gamma (PPARγ) agonist, rosiglitazone (thiazolidinedione), is a popular drug for treating diabetes; however, it leads to increased cardiovascular risk. Selective sodium-glucose co-transporter-2 (SGLT-2) inhibitor empagliflozin (EMPA) significantly improves endothelial dysfunction and mortality occurring through redox-dependent mechanisms. Although endothelial dysfunction and oxidative stress are alleviated by either metformin or EMPA, currently used drugs to treat obesity-related diabetes neither possess the same anti-inflammatory potential nor simultaneously target endothelial cell dysfunction and obesity equally. While therapeutic interventions with glucagon-like peptide-1 (GLP-1) receptor agonist liraglutide or bariatric surgery reverse regenerative cell exhaustion, support vascular repair mechanisms, and improve cardiometabolic risk in individuals with T2D and obesity, the GLP-1 analog exendin-4 attenuates endothelial endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
208
|
Engin A. Reappraisal of Adipose Tissue Inflammation in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:297-327. [PMID: 39287856 DOI: 10.1007/978-3-031-63657-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Chronic low-grade inflammation is a central component in the pathogenesis of obesity-related expansion of adipose tissue and complications in other metabolic tissues. Five different signaling pathways are defined as dominant determinants of adipose tissue inflammation: These are increased circulating endotoxin due to dysregulation in the microbiota-gut-brain axis, systemic oxidative stress, macrophage accumulation, and adipocyte death. Finally, the nucleotide-binding and oligomerization domain (NOD) leucine-rich repeat family pyrin domain-containing 3 (NLRP3) inflammasome pathway is noted to be a key regulator of metabolic inflammation. The NLRP3 inflammasome and associated metabolic inflammation play an important role in the relationships among fatty acids and obesity. Several highly active molecules, including primarily leptin, resistin, adiponectin, visfatin, and classical cytokines, are abundantly released from adipocytes. The most important cytokines that are released by inflammatory cells infiltrating obese adipose tissue are tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), monocyte chemoattractant protein 1 (MCP-1) (CCL-2), and IL-1. All these molecules mentioned above act on immune cells, causing local and then general inflammation. Three metabolic pathways are noteworthy in the development of adipose tissue inflammation: toll-like receptor 4 (TLR4)/phosphatidylinositol-3'-kinase (PI3K)/Protein kinase B (Akt) signaling pathway, endoplasmic reticulum (ER) stress-derived unfolded protein response (UPR), and inhibitor of nuclear factor kappa-B kinase beta (IKKβ)-nuclear factor kappa B (NF-κB) pathway. In fact, adipose tissue inflammation is an adaptive response that contributes to a visceral depot barrier that effectively filters gut-derived endotoxin. Excessive fatty acid release worsens adipose tissue inflammation and contributes to insulin resistance. However, suppression of adipose inflammation in obesity with anti-inflammatory drugs is not a rational solution and paradoxically promotes insulin resistance, despite beneficial effects on weight gain. Inflammatory pathways in adipocytes are indeed indispensable for maintaining systemic insulin sensitivity. Cannabinoid type 1 receptor (CB1R) is important in obesity-induced pro-inflammatory response; however, blockade of CB1R, contrary to anti-inflammatory drugs, breaks the links between insulin resistance and adipose tissue inflammation. Obesity, however, could be decreased by improving leptin signaling, white adipose tissue browning, gut microbiota interactions, and alleviating inflammation. Furthermore, capsaicin synthesized by chilies is thought to be a new and promising therapeutic option in obesity, as it prevents metabolic endotoxemia and systemic chronic low-grade inflammation caused by high-fat diet.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
209
|
Engin AB. Message Transmission Between Adipocyte and Macrophage in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:273-295. [PMID: 39287855 DOI: 10.1007/978-3-031-63657-8_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Obesity is characterized by the chronic low-grade activation of the innate immune system. In this respect, macrophage-elicited metabolic inflammation and adipocyte-macrophage interaction have primary importance in obesity. Large quantity of macrophages is accumulated by different mechanisms in obese adipose tissue. Hypertrophic adipocyte-derived chemotactic monocyte chemoattractant protein-1 (MCP-1)/C-C chemokine receptor 2 (CCR2) pathway promotes more macrophage accumulation into the obese adipose tissue. However, obesity-induced changes in adipose tissue macrophage density are mainly dependent on increases in the triple-positive cluster of differentiation (CD)11b+ F4/80+ CD11c+ adipose tissue macrophage subpopulation. As epigenetic regulators, microRNAs (miRNAs) are one of the most important mediators of obesity. miRNAs are expressed by adipocytes as well as macrophages and regulate inflammation with the expression of target genes. A paracrine loop involving free fatty acids and tumor necrosis factor-alpha (TNF-α) between adipocytes and macrophages establishes a vicious cycle that aggravates inflammatory changes in the adipose tissue. Adipocyte-specific caspase-1 and production of interleukin-1beta (IL-1β) by macrophages; both adipocyte and macrophage induction by toll-like receptor-4 (TLR4) through nuclear factor-kappaB (NF-κB) activation; free fatty acid-induced and TLR-mediated activation of c-Jun N-terminal kinase (JNK)-related pro-inflammatory pathways in CD11c+ immune cells; are effective in mutual message transmission between adipocyte and macrophage and in the development of adipose tissue inflammation. Thus, the metabolic status of adipocytes and their released exosomes are important determinants of macrophage inflammatory output. However, old adipocytes are removed by macrophages through trogocytosis or sending an "eat me" signal. As a single miRNA can be able to regulate a variety of target genes and signaling pathways, reciprocal transfer of miRNAs between adipocytes and macrophages via miRNA-loaded exosomes reorganizes the different stages of obesity. Changes in the expression of circulating miRNAs because of obesity progression or anti-obesity treatment indicate that miRNAs could be used as potential biomarkers. Therefore, it is believed that targeting macrophage-associated miRNAs with anti-obesity miRNA-loaded nano-carriers may be successful in the attenuation of both obesity and adipose tissue inflammation in clinical practice. Moreover, miRNA-containing exosomes and transferable mitochondria between the adipocyte and macrophage are investigated as new therapeutic targets for obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| |
Collapse
|
210
|
Engin A. Lipid Storage, Lipolysis, and Lipotoxicity in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:97-129. [PMID: 39287850 DOI: 10.1007/978-3-031-63657-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The ratio of free fatty acid (FFA) turnover decreases significantly with the expansion of white adipose tissue. Adipose tissue and dietary saturated fatty acid levels significantly correlate with an increase in fat cell size and number. The G0/G1 switch gene 2 increases lipid content in adipocytes and promotes adipocyte hypertrophy through the restriction of triglyceride (triacylglycerol: TAG) turnover. Hypoxia in obese adipose tissue due to hypertrophic adipocytes results in excess deposition of extracellular matrix (ECM) components. Cluster of differentiation (CD) 44, as the main receptor of the extracellular matrix component regulates cell-cell and cell-matrix interactions including diet-induced insulin resistance. Excess TAGs, sterols, and sterol esters are surrounded by the phospholipid monolayer surface and form lipid droplets (LDs). Once LDs are formed, they grow up because of the excessive amount of intracellular FFA stored and reach a final size. The ratio of FFA turnover/lipolysis decreases significantly with increases in the degree of obesity. Dysfunctional adipose tissue is unable to expand further to store excess dietary lipids, increased fluxes of plasma FFAs lead to ectopic fatty acid deposition and lipotoxicity. Reduced neo-adipogenesis and dysfunctional lipid-overloaded adipocytes are hallmarks of hypertrophic obesity linked to insulin resistance. Obesity-associated adipocyte death exhibits feature of necrosis-like programmed cell death. Adipocyte death is a prerequisite for the transition from hypertrophic to hyperplastic obesity. Increased adipocyte number in obesity has life-long effects on white adipose tissue mass. The positive correlation between the adipose tissue volume and magnetic resonance imaging proton density fat fraction estimation is used for characterization of the obesity phenotype, as well as the risk stratification and selection of appropriate treatment strategies. In obese patients with type 2 diabetes, visceral adipocytes exposed to chronic/intermittent hyperglycemia develop a new microRNAs' (miRNAs') expression pattern. Visceral preadipocytes memorize the effect of hyperglycemia via changes in miRNAs' expression profile and contribute to the progression of diabetic phenotype. Nonsteroidal anti-inflammatory drugs, metformin, and statins can be beneficial in treating the local or systemic consequences of white adipose tissue inflammation. Rapamycin inhibits leptin-induced LD formation. Collectively, in this chapter, the concept of adipose tissue remodeling in response to adipocyte death or adipogenesis, and the complexity of LD interactions with the other cellular organelles are reviewed. Furthermore, clinical perspective of fat cell turnover in obesity is also debated.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
211
|
Engin A. Obesity-Associated Breast Cancer: Analysis of Risk Factors and Current Clinical Evaluation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:767-819. [PMID: 39287872 DOI: 10.1007/978-3-031-63657-8_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Several studies show that a significantly stronger association is obvious between increased body mass index (BMI) and higher breast cancer incidence. Additionally, obese and postmenopausal women are at higher risk of all-cause and breast cancer-specific mortality compared with non-obese women with breast cancer. In this context, increased levels of estrogens, excessive aromatization activity of the adipose tissue, overexpression of pro-inflammatory cytokines, insulin resistance, adipocyte-derived adipokines, hypercholesterolemia, and excessive oxidative stress contribute to the development of breast cancer in obese women. Genetic evaluation is an integral part of diagnosis and treatment for patients with breast cancer. Despite trimodality therapy, the four-year cumulative incidence of regional recurrence is significantly higher. Axillary lymph nodes as well as primary lesions have diagnostic, prognostic, and therapeutic significance for the management of breast cancer. In clinical setting, because of the obese population primary lesions and enlarged lymph nodes could be less palpable, the diagnosis may be challenging due to misinterpretation of physical findings. Thereby, a nomogram has been created as the "Breast Imaging Reporting and Data System" (BI-RADS) to increase agreement and decision-making consistency between mammography and ultrasonography (USG) experts. Additionally, the "breast density classification system," "artificial intelligence risk scores," ligand-targeted receptor probes," "digital breast tomosynthesis," "diffusion-weighted imaging," "18F-fluoro-2-deoxy-D-glucose positron emission tomography," and "dynamic contrast-enhanced magnetic resonance imaging (MRI)" are important techniques for the earlier detection of breast cancers and to reduce false-positive results. A high concordance between estrogen receptor (ER) and progesterone receptor (PR) status evaluated in preoperative percutaneous core needle biopsy and surgical specimens is demonstrated. Breast cancer surgery has become increasingly conservative; however, mastectomy may be combined with any axillary procedures, such as sentinel lymph node biopsy (SLNB) and/or axillary lymph node dissection whenever is required. As a rule, SLNB-guided axillary dissection in breast cancer patients who have clinically axillary lymph node-positive to node-negative conversion following neoadjuvant chemotherapy is recommended, because lymphedema is the most debilitating complication after any axillary surgery. There is no clear consensus on the optimal treatment of occult breast cancer, which is much discussed today. Similarly, the current trend in metastatic breast cancer is that the main palliative treatment option is systemic therapy.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
212
|
Carobbio S, Pellegrinelli V, Vidal-Puig A. Adipose Tissue Dysfunction Determines Lipotoxicity and Triggers the Metabolic Syndrome: Current Challenges and Clinical Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:231-272. [PMID: 39287854 DOI: 10.1007/978-3-031-63657-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The adipose tissue organ is organised as distinct anatomical depots located all along the body axis, and it is constituted of three different types of adipocytes: white, beige and brown, which are integrated with vascular, immune, neural, and extracellular stroma cells. These distinct adipocytes serve different specialised functions. The main function of white adipocytes is to ensure healthy storage of excess nutrients/energy and its rapid mobilisation to supply the demand of energy imposed by physiological cues in other organs, whereas brown and beige adipocytes are designed for heat production through uncoupling lipid oxidation from energy production. The concerted action of the three types of adipocytes/tissues ensures an optimal metabolic status. However, when one or several of these adipose depots become dysfunctional because of sustained lipid/nutrient overload, then insulin resistance and associated metabolic complications ensue. These metabolic alterations close a vicious cycle that negatively affects the adipose tissue functionality and compromises global metabolic homeostasis. Optimising white adipose tissue expandability and ensuring its functional metabolic flexibility and/or promoting brown/beige mediated thermogenic activity are complementary strategies that counteract obesity and its associated lipotoxic metabolic effects. However, the development of these therapeutic approaches requires a deep understanding of adipose tissue in all broad aspects. In this chapter, we will discuss the characteristics of the different adipose tissue depots with respect to origins and precursors recruitment, plasticity, cellular composition, and expandability capacity potential as well as molecular and metabolic characteristic signatures in both physiological and pathophysiological conditions. Current antilipotoxic strategies for future clinical application are also discussed in this chapter.
Collapse
Affiliation(s)
- Stefania Carobbio
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| | - Vanessa Pellegrinelli
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| |
Collapse
|
213
|
Kodosaki E, Daniels-Morgan A, Hassan N, Webb R, Morris K, Kelly CM. Development and characterisation of mgTHP-1, a novel in vitro model for neural macrophages with microglial characteristics. Neurol Res 2024; 46:1-13. [PMID: 37935114 DOI: 10.1080/01616412.2023.2257422] [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: 11/01/2022] [Accepted: 04/23/2023] [Indexed: 11/09/2023]
Abstract
Neuroinflammation is primarily characterised by activation of the brain's resident macrophages - the microglia. However, other central nervous system (CNS) cells also contribute to this response, including the astrocytes and endothelial cells. In addition, there is infiltration into the CNS of peripherally derived immune cells. Together these cells mediate inflammation by the production of cytokines, chemokines, reactive oxygen species, and secondary messengers, and enacting of the appropriate response to those signals. However, deciphering the specific contributions of each cell type has been challenging. Studying CNS cell biology is often challenging, as the isolation of primary cells is not always feasible, and differentiation towards microglia-like cells is complex. Here, we demonstrate a novel method whereby THP-1 monocytic cells are differentiated into neural macrophage cells with microglia-like cell characteristics. The cells, designated mgTHP-1, show typical morphological and gene expression patterns of resident CNS macrophages and functionally respond to inflammatory stimuli by producing inflammatory cytokines. Furthermore, with the addition of Vicenin-2 (an anti-inflammatory flavonoid) such responses can be reversed. This novel cell model will allow further investigations, and hence insights, into the neuroinflammatory mechanisms associated with CNS diseases.
Collapse
Affiliation(s)
- E Kodosaki
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - A Daniels-Morgan
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - N Hassan
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - R Webb
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - K Morris
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - C M Kelly
- Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| |
Collapse
|
214
|
Wouters F, van der Hilst J, Bogie J. Lipids in inflammasome activation and autoinflammatory disorders. J Allergy Clin Immunol 2024; 153:1-11. [PMID: 37871669 DOI: 10.1016/j.jaci.2023.10.008] [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: 05/16/2023] [Revised: 09/06/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023]
Abstract
Autoinflammatory diseases (AIDs) are a group of rare monogenetic disorders characterized by recurrent episodes of fever and systemic inflammation. A major pathologic hallmark of AIDs is excessive inflammasome assembly and activation, often the result of gain-of-function mutations in genes encoding core inflammasome components, including pyrin and cryopyrin. Recent advances in lipidomics have revealed that dysregulated metabolism of lipids such as cholesterol and fatty acids, especially in innate immune cells, exerts complex effects on inflammasome activation and the pathogenesis of AIDs. In this review, we summarize and discuss the impact of lipids and their metabolism on inflammasome activation and the disease pathogenesis of the most common AIDs, including familial Mediterranean fever, cryopyrin-associated periodic syndromes, and mevalonate kinase deficiency. We postulate that lipids hold diagnostic value in AIDs and that dietary and pharmacologic intervention studies could represent a promising approach to attenuate inflammasome activation and AID progression.
Collapse
Affiliation(s)
- Flore Wouters
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Jeroen van der Hilst
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium; Department of Infectious Diseases and Immune Pathology, Jessa General Hospital and Limburg Clinical Research Center, Hasselt, Belgium
| | - Jeroen Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium; University MS Center Hasselt, Pelt, Belgium.
| |
Collapse
|
215
|
Wu D, Lan Y, Chen S, Ding X, Chen G, Wu C, Balmer L, Xu W, Wu S, Wang W. Combined effect of adiposity and elevated inflammation on incident type 2 diabetes: a prospective cohort study. Cardiovasc Diabetol 2023; 22:351. [PMID: 38124083 PMCID: PMC10734163 DOI: 10.1186/s12933-023-02067-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/14/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Adiposity and elevated inflammation are two hallmarks of hyperglycemia. However, it is unknown whether clustering of elevated inflammation and adiposity interact act on diabetogenesis and lead to a greater risk for incident type 2 diabetes (T2D). METHODS Adiposity was indicated by body mass index, waist circumference and ultrasonography-measured fatty liver degrees. Elevated inflammation was indicated as high-sensitivity C-reactive protein levels ≥ 2 mg/L. Time-to-event survival analyses were conducted to investigate the joint effect of adiposity and inflammation on incident T2D on both multiplicative and additive scales. RESULTS Among 82,172 non-diabetic participants from a prospective cohort in China, 14,278 T2D occurred over a median follow-up of 11 years. In the multivariable-adjusted model, elevated inflammation [1.12 (1.08‒1.16)] and adiposity [1.76 (1.69‒1.83) for overweight/obesity, 1.49 (1.44‒1.55) for central obesity, and 2.02 (1.95‒2.09) for fatty liver] were significantly associated with incident diabetes. Higher adiposity-associated risks and incidence rates of diabetes were observed with elevated inflammation. When studying the joint effect, the adjusted HRs were 1.77 (1.69‒1.85) for overweight/obesity, 1.14 (1.06‒1.23) for elevated inflammation, and 2.08 (1.97‒2.19) for their joint effect, with a relative excess risk due to interaction of 0.17 (0.05‒0.28). The attributable proportions were 71.30% for overweight/obesity, 12.96% for elevated inflammation, and 15.74% for their interaction. Similar results were observed when adiposity was assessed as waist circumference or fatty liver. CONCLUSIONS Adiposity and elevated inflammation synergically lead to greater risks of incident diabetes than addition of each individual exposure. Strategies simultaneously targeting both risks should produce more benefits for diabetes prevention than through initiatives directed at each separate risk.
Collapse
Affiliation(s)
- Dan Wu
- Centre for Precision Health, Edith Cowan University School of Medical and Health Sciences, Room 521, Building 21/270 Joondalup Drive, Perth, WA, 6027, Australia
- Department of Pediatrics, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Jinping District, Shantou, 515041, Guangdong, China
| | - Yulong Lan
- Centre for Precision Health, Edith Cowan University School of Medical and Health Sciences, Room 521, Building 21/270 Joondalup Drive, Perth, WA, 6027, Australia
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Shuohua Chen
- Department of Cardiology, Kailuan General Hospital, Xinghua East Road, Tangshan, 063000, Hebei, China
| | - Xiong Ding
- School of Public Health, Wuhan University, Wuhan, China
| | | | - Chutao Wu
- Department of Emergency, Shantou Central Hospital, Shantou, Guangdong, China
| | - Lois Balmer
- Centre for Precision Health, Edith Cowan University School of Medical and Health Sciences, Room 521, Building 21/270 Joondalup Drive, Perth, WA, 6027, Australia
| | - Wencan Xu
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Jinping District, Shantou, 515041, Guangdong, China.
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, Xinghua East Road, Tangshan, 063000, Hebei, China.
| | - Wei Wang
- Centre for Precision Health, Edith Cowan University School of Medical and Health Sciences, Room 521, Building 21/270 Joondalup Drive, Perth, WA, 6027, Australia.
- Clinical Research Centre, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China.
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, Shandong, China.
| |
Collapse
|
216
|
Zhang Y, Yang W, Kumagai Y, Loza M, Zhang W, Park SJ, Nakai K. Multi-omics computational analysis unveils the involvement of AP-1 and CTCF in hysteresis of chromatin states during macrophage polarization. Front Immunol 2023; 14:1304778. [PMID: 38173717 PMCID: PMC10761412 DOI: 10.3389/fimmu.2023.1304778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
Macrophages display extreme plasticity, and the mechanisms and applications of polarization and de-/repolarization of macrophages have been extensively investigated. However, the regulation of macrophage hysteresis after de-/repolarization remains unclear. In this study, by using a large-scale computational analysis of macrophage multi-omics data, we report a list of hysteresis genes that maintain their expression patterns after polarization and de-/repolarization. While the polarization in M1 macrophages leads to a higher level of hysteresis in genes associated with cell cycle progression, cell migration, and enhancement of the immune response, we found weak levels of hysteresis after M2 polarization. During the polarization process from M0 to M1 and back to M0, the factors IRFs/STAT, AP-1, and CTCF regulate hysteresis by altering their binding sites to the chromatin. Overall, our results show that a history of polarization can lead to hysteresis in gene expression and chromatin accessibility over a given period. This study contributes to the understanding of de-/repolarization memory in macrophages.
Collapse
Affiliation(s)
- Yubo Zhang
- Department of Computational Biology and Medical Science, the University of Tokyo, Tokyo, Japan
| | - Wenbo Yang
- Department of Computational Biology and Medical Science, the University of Tokyo, Tokyo, Japan
| | - Yutaro Kumagai
- Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Martin Loza
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Weihang Zhang
- Department of Computational Biology and Medical Science, the University of Tokyo, Tokyo, Japan
| | - Sung-Joon Park
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Kenta Nakai
- Department of Computational Biology and Medical Science, the University of Tokyo, Tokyo, Japan
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
217
|
Kale D, Fatangare A, Phapale P, Sickmann A. Blood-Derived Lipid and Metabolite Biomarkers in Cardiovascular Research from Clinical Studies: A Recent Update. Cells 2023; 12:2796. [PMID: 38132115 PMCID: PMC10741540 DOI: 10.3390/cells12242796] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023] Open
Abstract
The primary prevention, early detection, and treatment of cardiovascular disease (CVD) have been long-standing scientific research goals worldwide. In the past decades, traditional blood lipid profiles have been routinely used in clinical practice to estimate the risk of CVDs such as atherosclerotic cardiovascular disease (ASCVD) and as treatment targets for the primary prevention of adverse cardiac events. These blood lipid panel tests often fail to fully predict all CVD risks and thus need to be improved. A comprehensive analysis of molecular species of lipids and metabolites (defined as lipidomics and metabolomics, respectively) can provide molecular insights into the pathophysiology of the disease and could serve as diagnostic and prognostic indicators of disease. Mass spectrometry (MS) and nuclear magnetic resonance (NMR)-based lipidomics and metabolomics analysis have been increasingly used to study the metabolic changes that occur during CVD pathogenesis. In this review, we provide an overview of various MS-based platforms and approaches that are commonly used in lipidomics and metabolomics workflows. This review summarizes the lipids and metabolites in human plasma/serum that have recently (from 2018 to December 2022) been identified as promising CVD biomarkers. In addition, this review describes the potential pathophysiological mechanisms associated with candidate CVD biomarkers. Future studies focused on these potential biomarkers and pathways will provide mechanistic clues of CVD pathogenesis and thus help with the risk assessment, diagnosis, and treatment of CVD.
Collapse
Affiliation(s)
- Dipali Kale
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany; (A.F.); (P.P.)
| | | | | | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany; (A.F.); (P.P.)
| |
Collapse
|
218
|
Mey JT, Vandagmansar B, Dantas WS, Belmont KP, Axelrod CL, Kirwan JP. Ketogenic propensity is differentially related to lipid-induced hepatic and peripheral insulin resistance. Acta Physiol (Oxf) 2023; 239:e14054. [PMID: 37840478 DOI: 10.1111/apha.14054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/11/2023] [Accepted: 09/26/2023] [Indexed: 10/17/2023]
Abstract
AIM Determine the ketogenic response (β-hydroxybutyrate, a surrogate of hepatic ketogenesis) to a controlled lipid overload in humans. METHODS In total, nineteen young, healthy adults (age: 28.4 ± 1.7 years; BMI: 22.7 ± 0.3 kg/m2 ) received either a 12 h overnight lipid infusion or saline in a randomized, crossover design. Plasma ketones and inflammatory markers were quantified by colorimetric and multiplex assays. Hepatic and peripheral insulin sensitivity was assessed by the hyperinsulinemic-euglycemic clamp. Skeletal muscle biopsies were obtained to quantify gene expression related to ketone body metabolism and inflammation. RESULTS By design, the lipid overload-induced hepatic (50%, p < 0.001) and peripheral insulin resistance (73%, p < 0.01) in healthy adults. Ketones increased with hyperlipidemia and were subsequently reduced with hyperinsulinemia during the clamp procedure (Saline: Basal = 0.22 mM, Insulin = 0.07 mM; Lipid: Basal = 0.78 mM, Insulin = 0.51 mM; 2-way ANOVA: Lipid p < 0.001, Insulin p < 0.001, Interaction p = 0.07). In the saline control condition, ketones did not correlate with hepatic or peripheral insulin sensitivity. Conversely, in the lipid condition, ketones were positively correlated with hepatic insulin sensitivity (r = 0.59, p < 0.01), but inversely related to peripheral insulin sensitivity (r = -0.64, p < 0.01). Hyperlipidemia increased plasma inflammatory markers, but did not impact skeletal muscle inflammatory gene expression. Gene expression related to ketone and fatty acid metabolism in skeletal muscle increased in response to hyperlipidemia. CONCLUSION This work provides important insight into the role of ketones in human health and suggests that ketone body metabolism is altered at the onset of lipid-induced insulin resistance.
Collapse
Affiliation(s)
- J T Mey
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - B Vandagmansar
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - W S Dantas
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - K P Belmont
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - C L Axelrod
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - J P Kirwan
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| |
Collapse
|
219
|
Ye T, Tao WY, Chen XY, Jiang C, Di B, Xu LL. Mechanisms of NLRP3 inflammasome activation and the development of peptide inhibitors. Cytokine Growth Factor Rev 2023; 74:1-13. [PMID: 37821254 DOI: 10.1016/j.cytogfr.2023.09.007] [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: 09/22/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023]
Abstract
The Nucleotide-binding domain leucine-rich repeat and pyrin domain containing receptor 3 (NLRP3), a member of the nucleotide-binding oligomerization domain (NOD) like receptors (NLRs) family, plays an important role in the innate immune response against pathogen invasions. NLRP3 inflammasome consisting of NLRP3 protein, the adapter protein apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD) (ASC), and the effector protein pro-caspase-1, is central to this process. Upon activation, NLRP3 inflammasome initiates the release of inflammatory cytokines and triggers a form of cell death known as pyroptosis. Dysregulation or inappropriate activation of NLRP3 has been implicated in various human diseases, including type 2 diabetes, colitis, depression, and gout. Consequently, understanding the mechanism underlying NLRP3 inflammasome activation is critical for the development of therapeutic drugs. In the pursuit of potential therapeutic agents, peptides present several advantages over small molecules. They offer higher selectivity, increased potency, reduced toxicity, and fewer off-target effects. The advancements in molecular biology have expanded the opportunities for applying peptides in medicine, unlocking their vast medical potential. This review begins by providing a comprehensive summary of recent research progress regarding the mechanisms governing NLRP3 inflammasome activation. Subsequently, we offer an overview of current peptide inhibitors capable of modulating the NLRP3 inflammasome activation pathway.
Collapse
Affiliation(s)
- Tao Ye
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Wei-Yan Tao
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Yi Chen
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Cheng Jiang
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| | - Bin Di
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| | - Li-Li Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
220
|
Zeng Y, Dong W, Zhang W, Deng B. Association of NLRPs with pathogenesis of dry age-related macular degeneration. Int Ophthalmol 2023; 43:4869-4878. [PMID: 37936001 DOI: 10.1007/s10792-023-02889-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 09/27/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Age-related macular degeneration (AMD) is the leading cause of blindness in the elderly population, and Dry AMD is the most common clinical subtype. However, effective measures for the early diagnosis and treatment of dry AMD have not been proposed. In recent years, NOD-like receptors (NLRs) have received attention in the study of AMD as an important class of pattern recognition receptors. We attempted to elucidate the pathogenesis of NLRs in dry AMD from the perspective of chronic inflammation. METHODS This study involved 13 patients with dry AMD, 10 age- and sex-matched normal population without any history of disease and 8 patients with wet AMD as controls. Using RT-qPCR, the mRNA expression levels of NLRs in peripheral blood peripheral blood mononuclear cells (PBMCs) were compared to analyze the statistical differences in the expression contents among the three populations. RESULTS The relative RNA expression of nucleotide-binding oligomerization-like receptor protein 12 (NLRP12) with negative regulation of inflammation was significantly lower in dry AMD patients than in normal people and wet AMD patients. And NLRX1, which also has an anti-inflammatory effect, was lower in dry AMD patients than in wet AMD patients. However, NLRP3 with proinflammatory effect was significantly expressed in wet AMD. CONCLUSION The significant decrease in NLRP12 in dry AMD may become a breakthrough in the study of dry AMD and systemic chronic inflammatory response. However, NLRP3 may have a more important role in wet AMD.
Collapse
Affiliation(s)
- Yiyun Zeng
- Department of Ophthalmology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 32# W. Sec 2, 1St Ring Rd., Chengdu, 611731, Sichuan, China
| | - Wentao Dong
- Department of Ophthalmology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 32# W. Sec 2, 1St Ring Rd., Chengdu, 611731, Sichuan, China
| | - Wanqiu Zhang
- Department of Ophthalmology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 32# W. Sec 2, 1St Ring Rd., Chengdu, 611731, Sichuan, China.
| | - Bolin Deng
- Department of Ophthalmology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 32# W. Sec 2, 1St Ring Rd., Chengdu, 611731, Sichuan, China.
| |
Collapse
|
221
|
Pérez-Gómez JM, Montero-Hidalgo AJ, Fuentes-Fayos AC, Sarmento-Cabral A, Guzmán-Ruiz R, Malagón MM, Herrera-Martínez AD, Gahete MD, Luque RM. Exploring the role of the inflammasomes on prostate cancer: Interplay with obesity. Rev Endocr Metab Disord 2023; 24:1165-1187. [PMID: 37819510 PMCID: PMC10697898 DOI: 10.1007/s11154-023-09838-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/21/2023] [Indexed: 10/13/2023]
Abstract
Obesity is a weight-related disorder characterized by excessive adipose tissue growth and dysfunction which leads to the onset of a systemic chronic low-grade inflammatory state. Likewise, inflammation is considered a classic cancer hallmark affecting several steps of carcinogenesis and tumor progression. In this regard, novel molecular complexes termed inflammasomes have been identified which are able to react to a wide spectrum of insults, impacting several metabolic-related disorders, but their contribution to cancer biology remains unclear. In this context, prostate cancer (PCa) has a markedly inflammatory component, and patients frequently are elderly individuals who exhibit weight-related disorders, being obesity the most prevalent condition. Therefore, inflammation, and specifically, inflammasome complexes, could be crucial players in the interplay between PCa and metabolic disorders. In this review, we will: 1) discuss the potential role of each inflammasome component (sensor, molecular adaptor, and targets) in PCa pathophysiology, placing special emphasis on IL-1β/NF-kB pathway and ROS and hypoxia influence; 2) explore the association between inflammasomes and obesity, and how these molecular complexes could act as the cornerstone between the obesity and PCa; and, 3) compile current clinical trials regarding inflammasome targeting, providing some insights about their potential use in the clinical practice.
Collapse
Affiliation(s)
- Jesús M Pérez-Gómez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Antonio J Montero-Hidalgo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - André Sarmento-Cabral
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Rocio Guzmán-Ruiz
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - María M Malagón
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Aura D Herrera-Martínez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Endocrinology and Nutrition Service, HURS/IMIBIC, Córdoba, Spain
| | - Manuel D Gahete
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain.
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain.
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain.
| |
Collapse
|
222
|
Klima ML, Kruger KA, Goldstein N, Pulido S, Low AYT, Assenmacher CA, Alhadeff AL, Betley JN. Anti-inflammatory effects of hunger are transmitted to the periphery via projection-specific AgRP circuits. Cell Rep 2023; 42:113338. [PMID: 37910501 DOI: 10.1016/j.celrep.2023.113338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 07/31/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023] Open
Abstract
Caloric restriction has anti-inflammatory effects. However, the coordinated physiological actions that lead to reduced inflammation in a state of caloric deficit (hunger) are largely unknown. Using a mouse model of injury-induced peripheral inflammation, we find that food deprivation reduces edema, temperature, and cytokine responses that occur after injury. The magnitude of the anti-inflammatory effect that occurs during hunger is more robust than that of non-steroidal anti-inflammatory drugs. The effects of hunger are recapitulated centrally by activity in nutrient-sensing hypothalamic agouti-related protein (AgRP)-expressing neurons. We find that AgRP neurons projecting to the paraventricular nucleus of the hypothalamus rapidly and robustly reduce inflammation and mediate the majority of hunger's anti-inflammatory effects. Intact vagal efferent signaling is required for the anti-inflammatory action of hunger, revealing a brain-to-periphery pathway for this reduction in inflammation. Taken together, these data begin to unravel a potent anti-inflammatory pathway engaged by hypothalamic AgRP neurons to reduce inflammation.
Collapse
Affiliation(s)
- Michelle L Klima
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kayla A Kruger
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nitsan Goldstein
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Santiago Pulido
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aloysius Y T Low
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charles-Antoine Assenmacher
- Comparative Pathology Core, Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Amber L Alhadeff
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA; Monell Chemical Senses Center, Philadelphia, PA 19104, USA.
| | - J Nicholas Betley
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
223
|
Pain P, Spinelli F, Gherardi G. Mitochondrial Cation Signalling in the Control of Inflammatory Processes. Int J Mol Sci 2023; 24:16724. [PMID: 38069047 PMCID: PMC10706693 DOI: 10.3390/ijms242316724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/17/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Mitochondria are the bioenergetic organelles responsible for the maintenance of cellular homeostasis and have also been found to be associated with inflammation. They are necessary to induce and maintain innate and adaptive immune cell responses, acting as signalling platforms and mediators in effector responses. These organelles are also known to play a pivotal role in cation homeostasis as well, which regulates the inflammatory responses through the modulation of these cation channels. In particular, this review focuses on mitochondrial Ca2+ and K+ fluxes in the regulation of inflammatory response. Nevertheless, this review aims to understand the interplay of these inflammation inducers and pathophysiological conditions. In detail, we discuss some examples of chronic inflammation such as lung, bowel, and metabolic inflammatory diseases caused by a persistent activation of the innate immune response due to a dysregulation of mitochondrial cation homeostasis.
Collapse
Affiliation(s)
| | | | - Gaia Gherardi
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (P.P.); (F.S.)
| |
Collapse
|
224
|
Cheng X, Jiang S, Pan B, Xie W, Meng J. Ectopic and visceral fat deposition in aging, obesity, and idiopathic pulmonary fibrosis: an interconnected role. Lipids Health Dis 2023; 22:201. [PMID: 38001499 PMCID: PMC10668383 DOI: 10.1186/s12944-023-01964-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is considered an age-related disease. Age-related changes, along with other factors such as obesity, hormonal imbalances, and various metabolic disorders, lead to ectopic fat deposition (EFD). This accumulation of fat outside of its normal storage sites is associated with detrimental effects such as lipotoxicity, oxidative stress, inflammation, and insulin resistance. This narrative review provides an overview of the connection between ectopic and visceral fat deposition in aging, obesity, and IPF. It also elucidates the mechanism by which ectopic fat deposition in the airways and lungs, pericardium, skeletal muscles, and pancreas contributes to lung injury and fibrosis in patients with IPF, directly or indirectly. Moreover, the review discusses the impact of EFD on the severity of the disease, quality of life, presence of comorbidities, and overall prognosis in IPF patients. The review provides detailed information on recent research regarding representative lipid-lowering drugs, hypoglycemic drugs, and lipid-targeting drugs in animal experiments and clinical studies. This may offer new therapeutic directions for patients with IPF.
Collapse
Affiliation(s)
- Xiaoyun Cheng
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Tongzipo Road 138, Yuelu District, Changsha, 410000, China
- Hunan Key Laboratory of Organ Fibrosis, Tongzipo Road 138, Yuelu District, Changsha, 410000, China
| | - Shuhan Jiang
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Tongzipo Road 138, Yuelu District, Changsha, 410000, China
- Hunan Key Laboratory of Organ Fibrosis, Tongzipo Road 138, Yuelu District, Changsha, 410000, China
| | - Boyu Pan
- Departments of Orthopedics, The Third Hospital of Changsha, Laodong West Road 176, Tianxin District, Changsha, 410000, China
| | - Wei Xie
- Department of Cardiology, Xiangya Hospital of Central South University, Furong Middle Road 36, Kaifu District, Changsha, 410000, China
| | - Jie Meng
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Tongzipo Road 138, Yuelu District, Changsha, 410000, China.
- Hunan Key Laboratory of Organ Fibrosis, Tongzipo Road 138, Yuelu District, Changsha, 410000, China.
| |
Collapse
|
225
|
Schleh MW, Caslin HL, Garcia JN, Mashayekhi M, Srivastava G, Bradley AB, Hasty AH. Metaflammation in obesity and its therapeutic targeting. Sci Transl Med 2023; 15:eadf9382. [PMID: 37992150 PMCID: PMC10847980 DOI: 10.1126/scitranslmed.adf9382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 08/29/2023] [Indexed: 11/24/2023]
Abstract
Obesity-associated inflammation is a systemic process that affects all metabolic organs. Prominent among these is adipose tissue, where cells of the innate and adaptive immune system are markedly changed in obesity, implicating these cells in a range of processes linking immune memory to metabolic regulation. Furthermore, weight loss and weight cycling have unexpected effects on adipose tissue immune populations. Here, we review the current literature on the roles of various immune cells in lean and obese adipose tissue. Within this context, we discuss pharmacological and nonpharmacological approaches to obesity treatment and their impact on systemic inflammation.
Collapse
Affiliation(s)
- Michael W. Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Heather L. Caslin
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jamie N. Garcia
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Mona Mashayekhi
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Gitanjali Srivastava
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Weight Loss Center, Vanderbilt University Medical Center, Nashville, TN 37204 USA
| | - Anna B. Bradley
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Weight Loss Center, Vanderbilt University Medical Center, Nashville, TN 37204 USA
- VA Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Alyssa H. Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- VA Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| |
Collapse
|
226
|
Shi C, Zhang X, Chi X, Zhou YR, Lyu W, Gao T, Zhou J, Chen Y, Yi C, Sun X, Zhang L, Liu Z. Discovery of NLRP3 inhibitors using machine learning: Identification of a hit compound to treat NLRP3 activation-driven diseases. Eur J Med Chem 2023; 260:115784. [PMID: 37672931 DOI: 10.1016/j.ejmech.2023.115784] [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: 07/18/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/08/2023]
Abstract
NLRP3 is vital in developing many human diseases as one of the most critical inflammasomes. Developing related inhibitors has been instrumental in advancing the development of therapies for associated diseases. To date, there are no NLRP3 inhibitors on the market. This study identified a series of NLRP3 inhibitors using the self-developed machine learning model. Among them, CSC-6 was validated as the hit molecule with optimal activity and significantly inhibited IL-1β secreted by PMA-THP-1 cells (IC50 = 2.3 ± 0.38 μM). The results show that CSC-6 specifically binds NLRP3 and inhibits NLRP3 activation by blocking ASC oligomerization during NLRP3 assembly. In vivo experiments have demonstrated that CSC-6 effectively reduces the symptoms of NLRP3 overactivation-mediated sepsis and Gout in mouse models. Importantly, CSC-6 has lower cytotoxicity and exhibits better stability in human-derived liver microsomes, which is more favorable for the drug to maintain its efficacy in vivo for longer. The discovery of CSC-6 may contribute to the design and discovery of related NLRP3 inhibitors.
Collapse
Affiliation(s)
- Cheng Shi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Xiangyu Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Xiaowei Chi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Yi Ran Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Weiping Lyu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Tongfei Gao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Jiaxu Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Yanming Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Chuxiao Yi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Xiaojiao Sun
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
227
|
Kholodenko IV, Yarygin KN. Hepatic Macrophages as Targets for the MSC-Based Cell Therapy in Non-Alcoholic Steatohepatitis. Biomedicines 2023; 11:3056. [PMID: 38002056 PMCID: PMC10669188 DOI: 10.3390/biomedicines11113056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a serious public health issue associated with the obesity pandemic. Obesity is the main risk factor for the non-alcoholic fatty liver disease (NAFLD), which progresses to NASH and then to end-stage liver disease. Currently, there are no specific pharmacotherapies of NAFLD/NASH approved by the FDA or other national regulatory bodies and the treatment includes lifestyle adjustment and medicines for improving lipid metabolism, enhancing sensitivity to insulin, balancing oxidation, and counteracting fibrosis. Accordingly, further basic research and development of new therapeutic approaches are greatly needed. Mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles prevent induced hepatocyte death in vitro and attenuate NASH symptoms in animal models of the disease. They interact with hepatocytes directly, but also target other liver cells, including Kupffer cells and macrophages recruited from the blood flow. This review provides an update on the pathogenesis of NAFLD/NASH and the key role of macrophages in the development of the disease. We examine in detail the mechanisms of the cross-talk between the MSCs and the macrophages, which are likely to be among the key targets of MSCs and their derivatives in the course of NAFLD/NASH cell therapy.
Collapse
Affiliation(s)
- Irina V. Kholodenko
- Laboratory of Cell Biology, Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| | | |
Collapse
|
228
|
Liu J, Yao C, Wang Y, Zhao J, Luo H. Non-drug interventions of traditional Chinese medicine in preventing type 2 diabetes: a review. Chin Med 2023; 18:151. [PMID: 37964315 PMCID: PMC10644617 DOI: 10.1186/s13020-023-00854-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/23/2023] [Indexed: 11/16/2023] Open
Abstract
Traditional Chinese medicine (TCM) is increasingly used to manage type 2 diabetes and its nonpharmacological interventions are showing potential for preventing type 2 diabetes. This study mainly reviews relevant research. The most mentioned non-drug treatments for preventing type 2 diabetes in TCM are healthy diet, physical activity, emotional therapy, and acupuncture. In most studies, blood glucose status in patients with prediabetes and type 2 diabetes was significantly improved after TCM non-drug interventions, and there was no significant difference between the adverse effect of TCM and control groups or other intervention groups, while the methodological quality of the clinical trials involving TCM generally kept a low level. The effectiveness of TCM in preventing type 2 diabetes has yet to be validated in large randomized controlled trials and the underlying mechanism also needs further exploration.
Collapse
Affiliation(s)
- Jingying Liu
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, People's Republic of China
| | - Chun Yao
- Guangxi University of Chinese Medicine, Nanning, 530001, People's Republic of China
| | - Yitao Wang
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, People's Republic of China
| | - Jinmin Zhao
- College of Pharmacy, Guangxi Medical University, Nanning, 530021, People's Republic of China.
| | - Hua Luo
- Macau Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, People's Republic of China.
- College of Pharmacy, Guangxi Medical University, Nanning, 530021, People's Republic of China.
| |
Collapse
|
229
|
Ruck L, Wiegand S, Kühnen P. Relevance and consequence of chronic inflammation for obesity development. Mol Cell Pediatr 2023; 10:16. [PMID: 37957462 PMCID: PMC10643747 DOI: 10.1186/s40348-023-00170-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Increasing prevalence of morbid obesity accompanied by comorbidities like type 2 diabetes mellitus (T2DM) led to a demand for improving therapeutic strategies and pharmacological intervention options. Apart from genetics, inflammation processes have been hypothesized to be of importance for the development of obesity and related aspects like insulin resistance. MAIN TEXT Within this review, we provide an overview of the intricate interplay between chronic inflammation of the adipose tissue and the hypothalamus and the development of obesity. Further understanding of this relationship might improve the understanding of the underlying mechanism and may be of relevance for the establishment of new treatment strategies.
Collapse
Affiliation(s)
- Lisa Ruck
- Klinik Für Pädiatrische Endokrinologie und Diabetologie, Charité Universitätsmedizin, Berlin, Germany.
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Charitéplatz 1, 10117, Berlin, Germany.
| | - Susanna Wiegand
- Abteilung Interdisziplinär, Sozial-Pädiatrisches Zentrum, Charité Universitätsmedizin, Berlin, Germany
| | - Peter Kühnen
- Klinik Für Pädiatrische Endokrinologie und Diabetologie, Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
230
|
Wan T, Wang Y, He K, Zhu S. Microbial sensing in the intestine. Protein Cell 2023; 14:824-860. [PMID: 37191444 PMCID: PMC10636641 DOI: 10.1093/procel/pwad028] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/04/2023] [Indexed: 05/17/2023] Open
Abstract
The gut microbiota plays a key role in host health and disease, particularly through their interactions with the immune system. Intestinal homeostasis is dependent on the symbiotic relationships between the host and the diverse gut microbiota, which is influenced by the highly co-evolved immune-microbiota interactions. The first step of the interaction between the host and the gut microbiota is the sensing of the gut microbes by the host immune system. In this review, we describe the cells of the host immune system and the proteins that sense the components and metabolites of the gut microbes. We further highlight the essential roles of pattern recognition receptors (PRRs), the G protein-coupled receptors (GPCRs), aryl hydrocarbon receptor (AHR) and the nuclear receptors expressed in the intestinal epithelial cells (IECs) and the intestine-resident immune cells. We also discuss the mechanisms by which the disruption of microbial sensing because of genetic or environmental factors causes human diseases such as the inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Tingting Wan
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Yalong Wang
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Kaixin He
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Shu Zhu
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
- Department of Digestive Disease, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230001, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China
| |
Collapse
|
231
|
Wang J, Zhang S, Hu L, Wang Y, Liu K, Le J, Tan Y, Li T, Xue H, Wei Y, Zhong O, He J, Zi D, Lei X, Deng R, Luo Y, Tang M, Su M, Cao Y, Liu Q, Tang Z, Lei X. Pyrroloquinoline quinone inhibits PCSK9-NLRP3 mediated pyroptosis of Leydig cells in obese mice. Cell Death Dis 2023; 14:723. [PMID: 37935689 PMCID: PMC10630350 DOI: 10.1038/s41419-023-06162-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/25/2023] [Accepted: 09/19/2023] [Indexed: 11/09/2023]
Abstract
Abnormal lipid metabolism and chronic low-grade inflammation are the main traits of obesity. Especially, the molecular mechanism of concomitant deficiency in steroidogenesis-associated enzymes related to testosterone (T) synthesis of obesity dominated a decline in male fertility is still poorly understood. Here, we found that in vivo, supplementation of pyrroloquinoline quinone (PQQ) efficaciously ameliorated the abnormal lipid metabolism and testicular spermatogenic function from high-fat-diet (HFD)-induced obese mice. Moreover, the transcriptome analysis of the liver and testicular showed that PQQ supplementation not only inhibited the high expression of proprotein convertase subtilisin/Kexin type 9 (PCSK9) but also weakened the NOD-like receptor family pyrin domain containing 3 (NLRP3)-mediated pyroptosis, which both played a negative role in T synthesis of Leydig Cells (LCs). Eventually, the function and the pyroptosis of LCs cultured with palmitic acid in vitro were simultaneously benefited by suppressing the expression of NLRP3 or PCSK9 respectively, as well the parallel effects of PQQ were affirmed. Collectively, our data revealed that PQQ supplementation is a feasible approach to protect T synthesis from PCSK9-NLRP3 crosstalk-induced LCs' pyroptosis in obese men.
Collapse
Affiliation(s)
- Jinyuan Wang
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Shun Zhang
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Linlin Hu
- Reproductive Medicine Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Yan Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530004, China
| | - Ke Liu
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jianghua Le
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, China
| | - Yongpeng Tan
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Tianlong Li
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Haoxuan Xue
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yanhong Wei
- Reproductive Medicine Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Ou Zhong
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Junhui He
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Dan Zi
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xin Lei
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Renhe Deng
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yafei Luo
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Masong Tang
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Mingxuan Su
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yichang Cao
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Qingyou Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530004, China
| | - Zhihan Tang
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Xiaocan Lei
- Clinical Anatomy and Reproductive Medicine Application Institute, Department of Histology and Embryology, Postdoctoral Station for Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
232
|
Ramos-Tovar E, Muriel P. NLRP3 inflammasome in hepatic diseases: A pharmacological target. Biochem Pharmacol 2023; 217:115861. [PMID: 37863329 DOI: 10.1016/j.bcp.2023.115861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
The NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome pathway is mainly responsible for the activation and release of a cascade of proinflammatory mediators that contribute to the development of hepatic diseases. During alcoholic liver disease development, the NLRP3 inflammasome pathway contributes to the maturation of caspase-1, interleukin (IL)-1β, and IL-18, which induce a robust inflammatory response, leading to fibrosis by inducing profibrogenic hepatic stellate cell (HSC) activation. Substantial evidence demonstrates that nonalcoholic fatty liver disease (NAFLD) progresses to nonalcoholic steatohepatitis (NASH) via NLRP3 inflammasome activation, ultimately leading to fibrosis and hepatocellular carcinoma (HCC). Activation of the NLRP3 inflammasome in NASH can be attributed to several factors, such as reactive oxygen species (ROS), gut dysbiosis, leaky gut, which allow triggers such as cardiolipin, cholesterol crystals, endoplasmic reticulum stress, and uric acid to reach the liver. Because inflammation triggers HSC activation, the NLRP3 inflammasome pathway performs a central function in fibrogenesis regardless of the etiology. Chronic hepatic activation of the NLRP3 inflammasome can ultimately lead to HCC; however, inflammation also plays a role in decreasing tumor growth. Some data indicate that NLRP3 inflammasome activation plays an important role in autoimmune hepatitis, but the evidence is scarce. Most researchers have reported that NLRP3 inflammasome activation is essential in liver injury induced by a variety of drugs and hepatotropic virus infection; however, few reports indicate that this pathway can play a beneficial role by inducing liver regeneration. Modulation of the NLRP3 inflammasome appears to be a suitable strategy to treat liver diseases.
Collapse
Affiliation(s)
- Erika Ramos-Tovar
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina-IPN, Apartado Postal 11340, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México, México
| | - Pablo Muriel
- Laboratorio de Hepatología Experimental, Departamento de Farmacología, Cinvestav-IPN, Apartado Postal 14-740, Ciudad de México, México.
| |
Collapse
|
233
|
Li Y, Ji Y, Li F. A review: Mechanism and prospect of gastrodin in prevention and treatment of T2DM and COVID-19. Heliyon 2023; 9:e21218. [PMID: 37954278 PMCID: PMC10637887 DOI: 10.1016/j.heliyon.2023.e21218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 09/15/2023] [Accepted: 10/18/2023] [Indexed: 11/14/2023] Open
Abstract
Gastrodin is an extract from the dried tuber of the Chinese herb Gastrodia elata (Tian ma), with anti-inflammatory, antioxidant, and antiviral properties. Recent studies have shown that, compared to commonly used diabetes drugs, gastrodin has antidiabetic effects in multiple ways, with characteristics of low cost, high safety, less side effects, protection of β-cell function, relieving insulin resistance and alleviating multiple complications. In addition, it is confirmed that gastrodin can protect the function of lung and other organs, enhance antiviral activity via upregulating the type I interferon (IFN-I), and inhibit angiotensin II (AngII), a key factor in "cytokine storm" caused by COVID-19. Therefore, we reviewed the effect and mechanism of gastrodin on type 2 diabetes mellitus (T2DM), and speculated other potential mechanisms of gastrodin in alleviating insulin resistance from insulin signal pathway, inflammation, mitochondrial and endoplasmic reticulum and its potential in the prevention and treatment of COVID-19. We hope to provide new direction and treatment strategy for basic research and clinical work: gastrodin is considered as a drug for the prevention and treatment of diabetes and COVID-19.
Collapse
Affiliation(s)
- Yi Li
- Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Yuanyuan Ji
- Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Fenglan Li
- Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
234
|
Cavalcante-Silva J, Koh TJ. Targeting the NOD-Like Receptor Pyrin Domain Containing 3 Inflammasome to Improve Healing of Diabetic Wounds. Adv Wound Care (New Rochelle) 2023; 12:644-656. [PMID: 34841901 PMCID: PMC10701516 DOI: 10.1089/wound.2021.0148] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/08/2021] [Indexed: 12/21/2022] Open
Abstract
Significance: Chronic skin wounds are a significant health problem around the world, often leading to amputation and even death. Although persistent inflammation is a hallmark of these poorly healing wounds, few available therapies have been designed to target inflammation. In this review, we summarize available evidence of the role of the NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome in impaired wound healing and describe strategies to inhibit the inflammasome to improve wound healing. Recent Advances: The NLRP3 inflammasome plays an important physiological role in skin wound healing, during which transient inflammasome activity contributes to both epidermal and dermal healing. In contrast, sustained activity of the NLRP3 inflammasome leads to impaired epidermal and dermal healing associated with diabetes. Of importance, preclinical studies have demonstrated that inhibiting the NLRP3 inflammasome-induced resolution of inflammation, increased granulation tissue formation and collagen deposition, and accelerated reepithelialization and wound closure. Critical Issues: NLRP3 inflammasome inhibitors have appealing potential for translation into therapies for chronic wounds. Although preclinical studies have shown promising results, there is a need for human/clinical studies to evaluate dosing formulations, potential therapeutic effects, dose-response relationships, and possible side effects. Future Directions: Among strategies to inhibit the NLRP3 inflammasome, glyburide, metformin, peroxisome proliferator-activated receptor agonists, and the dipeptidyl peptidase 4 inhibitor saxagliptin appear to be closest to clinical translation, as these drugs are already Food and Drug Administration approved for other indications. Future clinical studies are needed to develop topical formulations of these drugs, and to assess the safety and efficacy of these inhibitors, to improve healing of chronic wounds.
Collapse
Affiliation(s)
- Jacqueline Cavalcante-Silva
- Center for Wound Healing and Tissue Regeneration; University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Kinesiology and Nutrition; University of Illinois at Chicago, Chicago, Illinois, USA
| | - Timothy J. Koh
- Center for Wound Healing and Tissue Regeneration; University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Kinesiology and Nutrition; University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
235
|
Bissonnette S, Lamantia V, Ouimet B, Cyr Y, Devaux M, Rabasa-Lhoret R, Chrétien M, Saleh M, Faraj M. Native low-density lipoproteins are priming signals of the NLRP3 inflammasome/interleukin-1β pathway in human adipose tissue and macrophages. Sci Rep 2023; 13:18848. [PMID: 37914804 PMCID: PMC10620147 DOI: 10.1038/s41598-023-45870-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023] Open
Abstract
Elevated plasma numbers of atherogenic apoB-lipoproteins (apoB), mostly as low-density lipoproteins (LDL), predict diabetes risk by unclear mechanisms. Upregulation of the NLRP3 inflammasome/interleukin-1 beta (IL-1β) system in white adipose tissue (WAT) is implicated in type 2 diabetes (T2D); however, metabolic signals that stimulate it remain unexplored. We hypothesized that (1) subjects with high-apoB have higher WAT IL-1β-secretion than subjects with low-apoB, (2) WAT IL-1β-secretion is associated with T2D risk factors, and (3) LDL prime and/or activate the WAT NLRP3 inflammasome. Forty non-diabetic subjects were assessed for T2D risk factors related to systemic and WAT glucose and fat metabolism. Regulation of the NLRP3 inflammasome was explored using LDL without/with the inflammasome's priming and activation controls (LPS and ATP). LDL induced IL1B-expression and IL-1β-secretion in the presence of ATP in WAT and macrophages. Subjects with high-apoB had higher WAT IL-1β-secretion independently of covariates. The direction of association of LDL-induced WAT IL-1β-secretion to T2D risk factors was consistently pathological in high-apoB subjects only. Adjustment for IL-1β-secretion eliminated the association of plasma apoB with T2D risk factors. In conclusion, subjects with high-apoB have higher WAT IL-1β-secretion that may explain their risk for T2D and may be related to LDL-induced priming of the NLRP3 inflammasome.ClinicalTrials.gov (NCT04496154): Omega-3 to Reduce Diabetes Risk in Subjects With High Number of Particles That Carry "Bad Cholesterol" in the Blood-Full Text View-ClinicalTrials.gov.
Collapse
Affiliation(s)
- Simon Bissonnette
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal (IRCM), Office 1770.2, 110, Avenue Des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Montréal Diabetes Research Center (MDRC), Montréal, QC, Canada
| | - Valérie Lamantia
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal (IRCM), Office 1770.2, 110, Avenue Des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Montréal Diabetes Research Center (MDRC), Montréal, QC, Canada
| | - Benjamin Ouimet
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal (IRCM), Office 1770.2, 110, Avenue Des Pins Ouest, Montréal, QC, H2W 1R7, Canada
| | - Yannick Cyr
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal (IRCM), Office 1770.2, 110, Avenue Des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Montréal Diabetes Research Center (MDRC), Montréal, QC, Canada
| | - Marie Devaux
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal (IRCM), Office 1770.2, 110, Avenue Des Pins Ouest, Montréal, QC, H2W 1R7, Canada
| | - Remi Rabasa-Lhoret
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal (IRCM), Office 1770.2, 110, Avenue Des Pins Ouest, Montréal, QC, H2W 1R7, Canada
- Montréal Diabetes Research Center (MDRC), Montréal, QC, Canada
| | - Michel Chrétien
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal (IRCM), Office 1770.2, 110, Avenue Des Pins Ouest, Montréal, QC, H2W 1R7, Canada
| | - Maya Saleh
- Faculty of Medicine, McGill University, Montréal, QC, Canada
- University of Bordeaux, Bordeaux, France
| | - May Faraj
- Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.
- Institut de Recherches Cliniques de Montréal (IRCM), Office 1770.2, 110, Avenue Des Pins Ouest, Montréal, QC, H2W 1R7, Canada.
- Montréal Diabetes Research Center (MDRC), Montréal, QC, Canada.
- Faculty of Medicine, McGill University, Montréal, QC, Canada.
| |
Collapse
|
236
|
Accogli T, Hibos C, Vegran F. Canonical and non-canonical functions of NLRP3. J Adv Res 2023; 53:137-151. [PMID: 36610670 PMCID: PMC10658328 DOI: 10.1016/j.jare.2023.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/22/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Since its discovery, NLRP3 is almost never separated from its major role in the protein complex it forms with ASC, NEK7 and Caspase-1, the inflammasome. This key component of the innate immune response mediates the secretion of proinflammatory cytokines IL-1β and IL-18 involved in immune response to microbial infection and cellular damage. However, NLRP3 has also other functions that do not involve the inflammasome assembly nor the innate immune response. These non-canonical functions have been poorly studied. Nevertheless, NLRP3 is associated with different kind of diseases probably through its inflammasome dependent function as through its inflammasome independent functions. AIM OF THE REVIEW The study and understanding of the canonical and non-canonical functions of NLRP3 can help to better understand its involvement in various pathologies. In parallel, the description of the mechanisms of action and regulation of its various functions, can allow the identification of new therapeutic strategies. KEY SCIENTIFIC CONCEPTS OF THE REVIEW NLRP3 functions have mainly been studied in the context of the inflammasome, in myeloid cells and in totally deficient transgenic mice. However, for several year, the work of different teams has proven that NLRP3 is also expressed in other cell types where it has functions that are independent of the inflammasome. If these studies suggest that NLRP3 could play different roles in the cytoplasm or the nucleus of the cells, the mechanisms underlying NLRP3 non-canonical functions remain unclear. This is why we propose in this review an inventory of the canonical and non-canonical functions of NLRP3 and their impact in different pathologies.
Collapse
Affiliation(s)
- Théo Accogli
- Faculté des Sciences de Santé- University of Burgundy, Dijon 21000, FRANCE; CAdIR Team - Centre de Recherche INSERM - UMR 1231, Dijon 21000, FRANCE
| | - Christophe Hibos
- Faculté des Sciences de Santé- University of Burgundy, Dijon 21000, FRANCE; CAdIR Team - Centre de Recherche INSERM - UMR 1231, Dijon 21000, FRANCE; Université de Bourgogne Franche-Comté, Dijon 21000, FRANCE
| | - Frédérique Vegran
- Faculté des Sciences de Santé- University of Burgundy, Dijon 21000, FRANCE; CAdIR Team - Centre de Recherche INSERM - UMR 1231, Dijon 21000, FRANCE; Department of Biology and Pathology of Tumors - Centre anticancéreux GF Leclerc, Dijon 21000, FRANCE.
| |
Collapse
|
237
|
Saleki K, Alijanizadeh P, Azadmehr A. Is neuropilin-1 the neuroimmune initiator of multi-system hyperinflammation in COVID-19? Biomed Pharmacother 2023; 167:115558. [PMID: 37748412 DOI: 10.1016/j.biopha.2023.115558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023] Open
Abstract
A major immunopathological feature of Coronavirus disease-2019 (COVID-19) is excessive inflammation in the form of "cytokine storm". The storm is characterized by injurious levels of cytokines which form a complicated network damaging different organs, including the lungs and the brain. The main starter of "cytokine network" hyperactivation in COVID-19 has not been discovered yet. Neuropilins (NRPs) are transmembrane proteins that act as neuronal guidance and angiogenesis modulators. The crucial function of NRPs in forming the nervous and vascular systems has been well-studied. NRP1 and NRP2 are the two identified homologs of NRP. NRP1 has been shown as a viral entry pathway for SARS-CoV2, which facilitates neuroinvasion by the virus within the central or peripheral nervous systems. These molecules directly interact with various COVID-19-related molecules, such as specific regions of the spike protein (major immune element of SARS-CoV2), vascular endothelial growth factor (VEGF) receptors, VEGFR1/2, and ANGPTL4 (regulator of vessel permeability and integrity). NRPs mainly play a role in hyperinflammatory injury of the CNS and lungs, and also the liver, kidney, pancreas, and heart in COVID-19 patients. New findings have suggested NRPs good candidates for pharmacotherapy of COVID-19. However, therapeutic targeting of NRP1 in COVID-19 is still in the preclinical phase. This review presents the implications of NRP1 in multi-organ inflammation-induced injury by SARS-CoV2 and provides insights for NRP1-targeting treatments for COVID-19 patients.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Department of e-Learning, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences(SBMU), Tehran, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Abbas Azadmehr
- Immunology Department, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
238
|
Sun W, Yue J, Xu T, Cui Y, Huang D, Shi H, Xiong J, Sun W, Yi Q. Xanthohumol alleviates palmitate-induced inflammation and prevents osteoarthritis progression by attenuating mitochondria dysfunction/NLRP3 inflammasome axis. Heliyon 2023; 9:e21282. [PMID: 37964828 PMCID: PMC10641167 DOI: 10.1016/j.heliyon.2023.e21282] [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] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent chronic degenerative joint disease worldwide. Obesity has been linked to OA, and increased free fatty acid levels (e.g., palmitate) contribute to inflammatory responses and cartilage degradation. Xanthohumol (Xn), a bioactive prenylated chalcone, was shown to exhibit antioxidative, anti-inflammatory, and anti-obesity capacities in multiple diseases. However, a clear description of the preventive effects of Xn on obesity-associated OA is unavailable. This study aimed to assess the chondroprotective function of Xn on obesity-related OA. The in vitro levels of inflammatory and ECM matrix markers in human chondrocytes were assessed after the chondrocytes were treated with PA and Xn. Additionally, in vivo cartilage degeneration was assessed following oral administration of HFD and Xn. This study found that Xn treatment completely reduces the inflammation and extracellular matrix degradation caused by PA. The proposed mechanism involves AMPK signaling pathway activation by Xn, which increases mitochondrial biogenesis, attenuates mitochondrial dysfunction, and inhibits NLRP3 inflammasome and the NF-κB signaling pathway induced by PA. In summary, this study highlights that Xn could decrease inflammation reactions and the degradation of the cartilage matrix induced by PA by inhibiting the NLRP3 inflammasome and attenuating mitochondria dysfunction in human chondrocytes.
Collapse
Affiliation(s)
- Weichao Sun
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, Guangdong, 518035, China
- The Central Laboratory, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, Guangdong 518035, China
| | - Jiaji Yue
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, Guangdong, 518035, China
| | - Tianhao Xu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, China
- Laboratory of Anesthesia and Organ Protection, Southwest Medical University, Luzhou, Sichuan, 646099, China
| | - Yinxing Cui
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, Guangdong, 518035, China
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Dixi Huang
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, Guangdong, 518035, China
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Houyin Shi
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, China
| | - Jianyi Xiong
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, Guangdong, 518035, China
| | - Wei Sun
- Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, Guangdong, 518035, China
| | - Qian Yi
- Laboratory of Anesthesia and Organ Protection, Southwest Medical University, Luzhou, Sichuan, 646099, China
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, Sichuan, 646000, China
| |
Collapse
|
239
|
Yang J, Liu S, Li Y, Fan Z, Meng Y, Zhou B, Zhang G, Zhan H. FABP4 in macrophages facilitates obesity-associated pancreatic cancer progression via the NLRP3/IL-1β axis. Cancer Lett 2023; 575:216403. [PMID: 37741433 DOI: 10.1016/j.canlet.2023.216403] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/25/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023]
Abstract
Obesity is an essential risk factor for pancreatic cancer (PC). Macrophage-induced inflammation plays a pivotal role in obesity-associated carcinogenesis and disease progression; however, the underlying molecular mechanisms remain unclear. In this study, we found that fatty acid-binding protein 4 (FABP4) overexpressed in serum of obese patients and was associated with poor overall survival. In vivo and in vitro experiments have revealed that FABP4 induces macrophage-related inflammation to promote cancer cell migration, invasion and metastasis under obese conditions. Mechanistically, FABP4 participates in transferring saturated fatty acid to induce macrophages pyroptosis in a caspase-1/GSDMD-dependent manner and mediates NOD-like receptor thermal protein domain associated protein 3 (NLRP3)/IL-1β axis in macrophages, which further regulates epithelial-mesenchymal transition signals to promote the migration, invasion, and metastasis of PC cells. Our results suggest that FABP4 in macrophages is a crucial regulator of the NLRP3/IL-1β axis to promote the progression of PC under obese conditions, which could act as a promising molecular target for treating of PC patients with obesity.
Collapse
Affiliation(s)
- Jian Yang
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Shujie Liu
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yongzheng Li
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yufan Meng
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Guangyong Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong Province, China.
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
240
|
Zhao Y, Yu S, Zhao H, Li L, Li Y, Liu M, Jiang L. Integrated multi-omics analysis reveals the positive leverage of citrus flavonoids on hindgut microbiota and host homeostasis by modulating sphingolipid metabolism in mid-lactation dairy cows consuming a high-starch diet. MICROBIOME 2023; 11:236. [PMID: 37880759 PMCID: PMC10598921 DOI: 10.1186/s40168-023-01661-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 09/03/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND Modern dairy diets have shifted from being forage-based to grain and energy dense. However, feeding high-starch diets can lead to a metabolic disturbance that is linked to dysregulation of the gastrointestinal microbiome and systemic inflammatory response. Plant flavonoids have recently attracted extensive interest due to their anti-inflammatory effects in humans and ruminants. Here, multi-omics analysis was conducted to characterize the biological function and mechanisms of citrus flavonoids in modulating the hindgut microbiome of dairy cows fed a high-starch diet. RESULTS Citrus flavonoid extract (CFE) significantly lowered serum concentrations of lipopolysaccharide (LPS) proinflammatory cytokines (TNF-α and IL-6), acute phase proteins (LPS-binding protein and haptoglobin) in dairy cows fed a high-starch diet. Dietary CFE supplementation increased fecal butyrate production and decreased fecal LPS. In addition, dietary CFE influenced the overall hindgut microbiota's structure and composition. Notably, potentially beneficial bacteria, including Bacteroides, Bifidobacterium, Alistipes, and Akkermansia, were enriched in CFE and were found to be positively correlated with fecal metabolites and host metabolites. Fecal and serum untargeted metabolomics indicated that CFE supplementation mainly emphasized the metabolic feature "sphingolipid metabolism." Metabolites associated with the sphingolipid metabolism pathway were positively associated with increased microorganisms in dairy cows fed CFE, particularly Bacteroides. Serum lipidomics analysis showed that the total contents of ceramide and sphingomyelin were decreased by CFE addition. Some differentially abundant sphingolipid species were markedly associated with serum IL-6, TNF-α, LPS, and fecal Bacteroides. Metaproteomics revealed that dietary supplementation with CFE strongly impacted the overall fecal bacterial protein profile and function. In CFE cows, enzymes involved in carbon metabolism, sphingolipid metabolism, and valine, leucine, and isoleucine biosynthesis were upregulated. CONCLUSIONS Our research indicates the importance of bacterial sphingolipids in maintaining hindgut symbiosis and homeostasis. Dietary supplementation with CFE can decrease systemic inflammation by maintaining hindgut microbiota homeostasis and regulating sphingolipid metabolism in dairy cows fed a high-starch diet. Video Abstract.
Collapse
Affiliation(s)
- Yuchao Zhao
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Shiqiang Yu
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Huiying Zhao
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Liuxue Li
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Yuqin Li
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Ming Liu
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Linshu Jiang
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China.
| |
Collapse
|
241
|
Ke P, Xu M, Feng J, Tian Q, He Y, Lu K, Lu Z. Association between weight change and risk of liver fibrosis in adults with type 2 diabetes. J Glob Health 2023; 13:04138. [PMID: 37856776 PMCID: PMC10586795 DOI: 10.7189/jogh.13.04138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Liver fibrosis plays a key role in the progression of non-alcoholic fatty liver disease to cirrhosis. Considering weight change is known to be closely associated with increased risk of liver fibrosis, we aimed to address a gap in evidence regarding the existence of this association in patients with type 2 diabetes (T2D). METHODS We included data on 622 T2D patients and 1618 non-T2D participants from the 2017-2018 cycle of the National Health and Nutrition Examination Survey (NHANES). We assessed liver fibrosis by the median values of liver stiffness measurement (LSM). According to the participants' body mass index (BMI) at age 25 (early adulthood), 10 years prior (middle adulthood), and at the 2017-2018 cycle (late adulthood), we categorised weight change patterns into stable non-obese, weight loss, weight gain, and stable obese. We applied logistic regression to association analysis and used population attributable fraction (PAF) to analyses hypothetical prevention regimens. RESULTS The prevalence of liver fibrosis was higher in T2D patients (23.04%) than in non-T2D participants (6.70%), while weight change was associated with a greater risk of fibrosis in the former compared to the latter group. Compared with T2D patients in the stable non-obese group, stable obese individuals from 10 years prior to the 2017-2018 cycle had the highest risk of developing liver fibrosis, corresponding to an adjusted odds ratio (aOR) of 3.13 (95% confidence interval = 1.84-5.48). Absolute weight change patterns showed that the risk of liver fibrosis was highest (aOR = 2.94) when T2D patients gained at least 20 kg of weight from 10 years prior to 2017-2018 cycle. CONCLUSIONS Obesity in middle and late adulthood is associated with an increased risk of T2D complicated with liver fibrosis.
Collapse
Affiliation(s)
- Pan Ke
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Minzhi Xu
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Feng
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qingfeng Tian
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yan He
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Kai Lu
- Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zuxun Lu
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
242
|
Li N, Zhang L, Wang X, Zhou Y, Gong L. Exploring exercise-driven inhibition of pyroptosis: novel insights into treating diabetes mellitus and its complications. Front Endocrinol (Lausanne) 2023; 14:1230646. [PMID: 37859981 PMCID: PMC10582706 DOI: 10.3389/fendo.2023.1230646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Diabetes mellitus (DM) and its complications are important, worldwide public health issues, exerting detrimental effects on human health and diminishing both quality of life and lifespan. Pyroptosis, as a new form of programmed cell death, plays a critical role in DM and its complications. Exercise has been shown to be an effective treatment for improving insulin sensitivity or preventing DM. However, the molecular mechanisms underlying the effects of exercise on pyroptosis-related diseases remain elusive. In this review, we provided a comprehensive elucidation of the molecular mechanisms underlying pyroptosis and the potential mechanism of exercise in the treatment of DM and its complications through the modulation of anti-pyroptosis-associated inflammasome pathways. Based on the existing evidence, further investigation into the mechanisms by which exercise inhibits pyroptosis through the regulation of inflammasome pathways holds promising potential for expanding preventive and therapeutic strategies for DM and facilitating the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Nan Li
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Liang Zhang
- School of Strength and Conditioning Training, Beijing Sport University, Beijing, China
| | - Xintang Wang
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Yue Zhou
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Lijing Gong
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China
| |
Collapse
|
243
|
Ryu S, Spadaro O, Sidorov S, Lee AH, Caprio S, Morrison C, Smith SR, Ravussin E, Shchukina I, Artyomov MN, Youm YH, Dixit VD. Reduction of SPARC protects mice against NLRP3 inflammasome activation and obesity. J Clin Invest 2023; 133:e169173. [PMID: 37781916 PMCID: PMC10541189 DOI: 10.1172/jci169173] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 08/01/2023] [Indexed: 10/03/2023] Open
Abstract
The comprehensive assessment of long-term effects of reducing intake of energy (CALERIE-II; NCT00427193) clinical trial established that caloric restriction (CR) in humans lowers inflammation. The identity and mechanism of endogenous CR-mimetics that can be deployed to control obesity-associated inflammation and diseases are not well understood. Our studies have found that 2 years of 14% sustained CR in humans inhibits the expression of the matricellular protein, secreted protein acidic and rich in cysteine (SPARC), in adipose tissue. In mice, adipose tissue remodeling caused by weight loss through CR and low-protein diet feeding decreased, while high-fat diet-induced (HFD-induced) obesity increased SPARC expression in adipose tissue. Inducible SPARC downregulation in adult mice mimicked CR's effects on lowering adiposity by regulating energy expenditure. Deletion of SPARC in adipocytes was sufficient to protect mice against HFD-induced adiposity, chronic inflammation, and metabolic dysfunction. Mechanistically, SPARC activates the NLRP3 inflammasome at the priming step and downregulation of SPARC lowers macrophage inflammation in adipose tissue, while excess SPARC activated macrophages via JNK signaling. Collectively, reduction of adipocyte-derived SPARC confers CR-like metabolic and antiinflammatory benefits in obesity by serving as an immunometabolic checkpoint of inflammation.
Collapse
Affiliation(s)
- Seungjin Ryu
- Department of Pathology and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pharmacology, College of Medicine, Hallym University, Chuncheon, Gangwon, South Korea
| | - Olga Spadaro
- Department of Pathology and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sviatoslav Sidorov
- Department of Pathology and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Aileen H. Lee
- Department of Pathology and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sonia Caprio
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Steven R. Smith
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, Florida, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Irina Shchukina
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Maxim N. Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yun-Hee Youm
- Department of Pathology and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Vishwa Deep Dixit
- Department of Pathology and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
- Yale Center for Research on Aging, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
244
|
Cho S, Ying F, Sweeney G. Sterile inflammation and the NLRP3 inflammasome in cardiometabolic disease. Biomed J 2023; 46:100624. [PMID: 37336361 PMCID: PMC10539878 DOI: 10.1016/j.bj.2023.100624] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/11/2023] [Accepted: 06/14/2023] [Indexed: 06/21/2023] Open
Abstract
Inflammation plays an important role in the pathophysiology of cardiometabolic diseases. Sterile inflammation, a non-infectious and damage-associated molecular pattern (DAMP)-induced innate response, is now well-established to be closely associated with development and progression of cardiometabolic diseases. The NOD-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome is well-established as a major player in sterile inflammatory responses. It is a multimeric cytosolic protein complex which regulates the activation of caspase-1 and subsequently promotes cleavage and release of interleukin (IL)-1 family cytokines, which have a deleterious impact on the development of cardiometabolic diseases. Therefore, targeting NLRP3 itself or the downstream consequences of NLRP3 activation represent excellent potential therapeutic targets in inflammatory cardiometabolic diseases. Here, we review our current understanding of the role which NLRP3 inflammasome regulation plays in cardiometabolic diseases such as obesity, diabetes, non-alcoholic steatohepatitis (NASH), atherosclerosis, ischemic heart disease and cardiomyopathy. Finally, we highlight the potential of targeting NLPR3 or related signaling molecules as a therapeutic approach.
Collapse
Affiliation(s)
- Sungji Cho
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Fan Ying
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, Ontario, Canada.
| |
Collapse
|
245
|
Wang A, Li Z, Sun Z, Zhang D, Ma X. Gut-derived short-chain fatty acids bridge cardiac and systemic metabolism and immunity in heart failure. J Nutr Biochem 2023; 120:109370. [PMID: 37245797 DOI: 10.1016/j.jnutbio.2023.109370] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 04/24/2023] [Accepted: 05/06/2023] [Indexed: 05/30/2023]
Abstract
Heart failure (HF) represents a group of complex clinical syndromes with high morbidity and mortality and has a significant global health burden. Inflammation and metabolic disorders are closely related to the development of HF, which are complex and depend on the severity and type of HF and common metabolic comorbidities such as obesity and diabetes. An increasing body of evidence indicates the importance of short-chain fatty acids (SCFAs) in regulating cardiac function. In addition, SCFAs represent a unique class of metabolites and play a distinct role in shaping systemic immunity and metabolism. In this review, we reveal the role of SCFAs as a link between metabolism and immunity, which regulate cardiac and systemic immune and metabolic systems by acting as energy substrates, inhibiting the expression of histone deacetylase (HDAC) regulated genes and activating G protein-coupled receptors (GPCRs) signaling. Ultimately cardiac efficiency is improved, cardiac inflammation alleviated and cardiac function in failing hearts enhanced. In conclusion, SCFAs represent a new therapeutic approach for HF.
Collapse
Affiliation(s)
- Anzhu Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhendong Li
- Qingdao West Coast New Area People's Hospital, Qingdao, China
| | - Zhuo Sun
- Qingdao West Coast New Area People's Hospital, Qingdao, China
| | - Dawu Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Xiaochang Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China.
| |
Collapse
|
246
|
Qiu T, Wu C, Yao X, Han Q, Wang N, Yuan W, Zhang J, Shi Y, Jiang L, Liu X, Yang G, Sun X. AS3MT facilitates NLRP3 inflammasome activation by m 6A modification during arsenic-induced hepatic insulin resistance. Cell Biol Toxicol 2023; 39:2165-2181. [PMID: 35226250 PMCID: PMC8882720 DOI: 10.1007/s10565-022-09703-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/16/2022] [Indexed: 01/01/2023]
Abstract
N6-methyladenosine (m6A) messenger RNA methylation is the most widespread gene regulatory mechanism affecting liver functions and disorders. However, the relationship between m6A methylation and arsenic-induced hepatic insulin resistance (IR), which is a critical initiating event in arsenic-induced metabolic syndromes such as type 2 diabetes (T2D) and non-alcoholic fatty liver disease (NAFLD), remains unclear. Here, we showed that arsenic treatment facilitated methyltransferase-like 14 (METTL14)-mediated m6A methylation, and that METTL14 interference reversed arsenic-impaired hepatic insulin sensitivity. We previously showed that arsenic-induced NOD-like receptor protein 3 (NLRP3) inflammasome activation contributed to hepatic IR. However, the regulatory mechanisms underlying the role of arsenic toward the post-transcriptional modification of NLRP3 remain unclear. Here, we showed that NLRP3 mRNA stability was enhanced by METTL14-mediated m6A methylation during arsenic-induced hepatic IR. Furthermore, we demonstrated that arsenite methyltransferase (AS3MT), an essential enzyme in arsenic metabolic processes, interacted with NLRP3 to activate the inflammasome, thereby contributing to arsenic-induced hepatic IR. Also, AS3MT strengthened the m6A methylase association with NLRP3 to stabilize m6A-modified NLRP3. In summary, we showed that AS3MT-induced m6A modification critically regulated NLRP3 inflammasome activation during arsenic-induced hepatic IR, and we identified a novel post-transcriptional function of AS3MT in promoting arsenicosis.
Collapse
Affiliation(s)
- Tianming Qiu
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Chenbing Wu
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Xiaofeng Yao
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Qiuyue Han
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Ningning Wang
- Department of Nutrition and Food Safety, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Weizhuo Yuan
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Jingyuan Zhang
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Yan Shi
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Liping Jiang
- Preventive Medicine Laboratory, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Xiaofang Liu
- Department of Nutrition and Food Safety, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Guang Yang
- Department of Nutrition and Food Safety, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Xiance Sun
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China.
- Global Health Research Center, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China.
| |
Collapse
|
247
|
Mandal S, Jaganathan S, Kondal D, Schwartz JD, Tandon N, Mohan V, Prabhakaran D, Narayan KMV. PM 2.5 exposure, glycemic markers and incidence of type 2 diabetes in two large Indian cities. BMJ Open Diabetes Res Care 2023; 11:e003333. [PMID: 37797962 PMCID: PMC10565186 DOI: 10.1136/bmjdrc-2023-003333] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023] Open
Abstract
INTRODUCTION Exposure to fine particulate matter has been associated with several cardiovascular and cardiometabolic diseases. However, such evidence mostly originates from low-pollution settings or cross-sectional studies, thus necessitating evidence from regions with high air pollution levels, such as India, where the burden of non-communicable diseases is high. RESEARCH DESIGN AND METHODS We studied the associations between ambient PM2.5 levels and fasting plasma glucose (FPG), glycosylated hemoglobin (HbA1c) and incident type 2 diabetes mellitus (T2DM) among 12 064 participants in an adult cohort from urban Chennai and Delhi, India. A meta-analytic approach was used to combine estimates, obtained from mixed-effects models and proportional hazards models, from the two cities. RESULTS We observed that 10 μg/m3 differences in monthly average exposure to PM2.5 was associated with a 0.40 mg/dL increase in FPG (95% CI 0.22 to 0.58) and 0.021 unit increase in HbA1c (95% CI 0.009 to 0.032). Further, 10 μg/m3 differences in annual average PM2.5 was associated with 1.22 (95% CI 1.09 to 1.36) times increased risk of incident T2DM, with non-linear exposure response. CONCLUSIONS We observed evidence of temporal association between PM2.5 exposure, and higher FPG and incident T2DM in two urban environments in India, thus highlighting the potential for population-based mitigation policies to reduce the growing burden of diabetes.
Collapse
Affiliation(s)
| | | | - Dimple Kondal
- Centre for Chronic Disease Control, New Delhi, India
- Public Health Foundation of India, New Delhi, Delhi, India
| | - Joel D Schwartz
- Harvard T H Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Nikhil Tandon
- Department of Endocrinology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Dorairaj Prabhakaran
- Centre for Chronic Disease Control, New Delhi, India
- Public Health Foundation of India, New Delhi, Delhi, India
| | - K M Venkat Narayan
- Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
248
|
Rodrigues E-Lacerda R, Fang H, Robin N, Bhatwa A, Marko DM, Schertzer JD. Microbiota and Nod-like receptors balance inflammation and metabolism during obesity and diabetes. Biomed J 2023; 46:100610. [PMID: 37263539 PMCID: PMC10505681 DOI: 10.1016/j.bj.2023.100610] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/03/2023] Open
Abstract
Gut microbiota influence host immunity and metabolism during obesity. Bacterial sensors of the innate immune system relay signals from specific bacterial components (i.e., postbiotics) that can have opposing outcomes on host metabolic inflammation. NOD-like receptors (NLRs) such as Nod1 and Nod2 both recruit receptor-interacting protein kinase 2 (RIPK2) but have opposite effects on blood glucose control. Nod1 connects bacterial cell wall-derived signals to metabolic inflammation and insulin resistance, whereas Nod2 can promote immune tolerance, insulin sensitivity, and better blood glucose control during obesity. NLR family pyrin domain containing (NLRP) inflammasomes can also generate divergent metabolic outcomes. NLRP1 protects against obesity and metabolic inflammation potentially because of a bias toward IL-18 regulation, whereas NLRP3 appears to have a bias toward IL-1β-mediated metabolic inflammation and insulin resistance. Targeting specific postbiotics that improve immunometabolism is a key goal. The Nod2 ligand, muramyl dipeptide (MDP) is a short-acting insulin sensitizer during obesity or during inflammatory lipopolysaccharide (LPS) stress. LPS with underacylated lipid-A antagonizes TLR4 and counteracts the metabolic effects of inflammatory LPS. Providing underacylated LPS derived from Rhodobacter sphaeroides improved insulin sensitivity in obese mice. Therefore, certain types of LPS can generate metabolically beneficial metabolic endotoxemia. Engaging protective adaptive immunoglobulin immune responses can also improve blood glucose during obesity. A bacterial vaccine approach using an extract of the entire bacterial community in the upper gut promotes protective adaptive immune response and long-lasting improvements in blood glucose control. A key future goal is to identify and combine postbiotics that cooperate to improve blood glucose control.
Collapse
Affiliation(s)
- Rodrigo Rodrigues E-Lacerda
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, And Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Han Fang
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, And Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Nazli Robin
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, And Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Arshpreet Bhatwa
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, And Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Daniel M Marko
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, And Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, And Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
249
|
Peixoto FC, Zanette DL, Cardoso TM, Nascimento MT, Sanches RCO, Aoki M, Scott P, Oliveira SC, Carvalho EM, Carvalho LP. Leishmania braziliensis exosomes activate human macrophages to produce proinflammatory mediators. Front Immunol 2023; 14:1256425. [PMID: 37841240 PMCID: PMC10569463 DOI: 10.3389/fimmu.2023.1256425] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/30/2023] [Indexed: 10/17/2023] Open
Abstract
Exosomes, organelles measuring 30-200nm, are secreted by various cell types. Leishmania exosomes consist of many proteins, including heat shock proteins, annexins, Glycoprotein 63, proteins exerting signaling activity and those containing mRNA and miRNA. Studies have demonstrated that Leishmania donovani exosomes downregulate IFN-γ and inhibit the expression of microbicidal molecules, such as TNF and nitric oxide, thus creating a microenvironment favoring parasite proliferation. Despite lacking immunological memory, data in the literature suggest that, following initial stimulation, mononuclear phagocytes may become "trained" to respond more effectively to subsequent stimuli. Here we characterized the effects of macrophage sensitization using L. braziliensis exosomes prior to infection by the same pathogen. Human macrophages were stimulated with L. braziliensis exosomes and then infected with L. braziliensis. Higher levels of IL-1β and IL-6 were detected in cultures sensitized prior to infection compared to unstimulated infected cells. Moreover, stimulation with L. braziliensis exosomes induced macrophage production of IL-1β, IL-6, IL-10 and TNF. Inhibition of exosome secretion by L. braziliensis prior to macrophage infection reduced cytokine production and produced lower infection rates than untreated infected cells. Exosome stimulation also induced the consumption/regulation of NLRP3 inflammasome components in macrophages, while the blockade of NLRP3 resulted in lower levels of IL-6 and IL-1β. Our results suggest that L. braziliensis exosomes stimulate macrophages, leading to an exacerbated inflammatory state that may be NLRP3-dependent.
Collapse
Affiliation(s)
- Fabio C. Peixoto
- Laboratório de Pesquisas Clínicas (LAPEC), Instituto Gonçalo Moniz (IGM), Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Programa de Pós Graduação em Ciências da Saúde, Universidade Federal da Bahia, UFBA, Salvador, Bahia, Brazil
| | - Dalila L. Zanette
- Laboratory for Applied Science and Technology in Health, Instituto Carlos Chagas – Oswaldo Cruz Foundation (FIOCRUZ) Paraná (ICC), Curitiba, Paraná, Brazil
| | - Thiago M. Cardoso
- Laboratório de Pesquisas Clínicas (LAPEC), Instituto Gonçalo Moniz (IGM), Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
| | - Mauricio T. Nascimento
- Laboratório de Pesquisas Clínicas (LAPEC), Instituto Gonçalo Moniz (IGM), Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Programa de Pós Graduação em Ciências da Saúde, Universidade Federal da Bahia, UFBA, Salvador, Bahia, Brazil
| | - Rodrigo C. O. Sanches
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mateus Aoki
- Laboratory for Applied Science and Technology in Health, Instituto Carlos Chagas – Oswaldo Cruz Foundation (FIOCRUZ) Paraná (ICC), Curitiba, Paraná, Brazil
| | - Phillip Scott
- University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA, United States
| | - Sérgio C. Oliveira
- Departamento de Imunologia, Instituto de Ciencias Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Edgar M. Carvalho
- Laboratório de Pesquisas Clínicas (LAPEC), Instituto Gonçalo Moniz (IGM), Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Programa de Pós Graduação em Ciências da Saúde, Universidade Federal da Bahia, UFBA, Salvador, Bahia, Brazil
- Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Lucas P. Carvalho
- Laboratório de Pesquisas Clínicas (LAPEC), Instituto Gonçalo Moniz (IGM), Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil
- Programa de Pós Graduação em Ciências da Saúde, Universidade Federal da Bahia, UFBA, Salvador, Bahia, Brazil
- Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| |
Collapse
|
250
|
Liu B, Wu Y, Liang T, Zhou Y, Chen G, He J, Ji C, Liu P, Zhang C, Lin J, Shi K, Luo Z, Liu N, Su X. Betulinic Acid Attenuates Osteoarthritis via Limiting NLRP3 Inflammasome Activation to Decrease Interleukin-1 β Maturation and Secretion. Mediators Inflamm 2023; 2023:3706421. [PMID: 37789884 PMCID: PMC10545461 DOI: 10.1155/2023/3706421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 07/09/2023] [Accepted: 08/01/2023] [Indexed: 10/05/2023] Open
Abstract
Introduction Osteoarthritis (OA) is the most common degenerative joint disorder. Prior studies revealed that activation of NLRP3 inflammasome could promote the activation and secretion of interleukin-1β (IL-1β), which has an adverse effect on the progression of OA. Betulinic acid (BA) is a compound extract of birch, whether it can protect against OA and the mechanisms involved are still unknown. Materials and Methods In vivo experiments, using gait analysis, ELISA, micro-CT, and scanning electron microscopy (SEM), histological staining, immunohistological (IHC) and immunofluorescence (IF) staining, and atomic force microscopy (AFM) to assess OA progression after intraperitoneal injection of 5 and 15 mg/kg BA in an OA mouse model. In vitro experiments, caspase-1, IL-1β, and the N-terminal fragment of gasdermin D (GSDMD-NT) were measured in bone marrow-derived macrophages (BMDMs) by using ELISA, western blot, and immunofluorescence staining. Results We demonstrated that OA progression can be postponed with intraperitoneal injection of 5 and 15 mg/kg BA in an OA mouse model. Specifically, BA postponed DMM-induced cartilage deterioration, alleviated subchondral bone sclerosis, and relieved synovial inflammation. In vitro studies, the activated NLRP3 inflammasome produces mature IL-1β by facilitating the cleavage of pro-IL-1β, and BA could inhibit the activation of NLRP3 inflammasome in BMDMs. Conclusions Taken together, our analyses revealed that BA attenuates OA via limiting NLRP3 inflammasome activation to decrease the IL-1β maturation and secretion.
Collapse
Affiliation(s)
- Bo Liu
- Department of Orthopaedics, People's Hospital of Leshan, 238 Baita Road, Leshan 614000, Sichuan, China
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
- Orthopaedic Institute, Soochow University, 708 Renmin Road, Suzhou 215006, Jiangsu, China
| | - Yanglin Wu
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
- Orthopaedic Institute, Soochow University, 708 Renmin Road, Suzhou 215006, Jiangsu, China
- Department of Orthopaedics, Tenth People's Hospital of Tongji University, 301 Middle Yanchang Road, Shanghai 200072, Shanghai, China
| | - Ting Liang
- Orthopaedic Institute, Soochow University, 708 Renmin Road, Suzhou 215006, Jiangsu, China
| | - Yunlong Zhou
- Department of Orthopaedics, People's Hospital of Leshan, 238 Baita Road, Leshan 614000, Sichuan, China
| | - Guangdong Chen
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Jiaheng He
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
- Orthopaedic Institute, Soochow University, 708 Renmin Road, Suzhou 215006, Jiangsu, China
- Department of Orthopaedics, Jiangsu Shengze Hospital, No. 1399, Market West Road, Shengze 215000, Jiangsu, China
| | - Chenchen Ji
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
- Orthopaedic Institute, Soochow University, 708 Renmin Road, Suzhou 215006, Jiangsu, China
- Stroke Intensive Care Unit, Children's Hospital of Soochow University, 92 Zhongnan Road, Suzhou 215006, Jiangsu, China
| | - Peixin Liu
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
- Orthopaedic Institute, Soochow University, 708 Renmin Road, Suzhou 215006, Jiangsu, China
- Department of Orthopedics, Suzhou Xiangcheng People's Hospital, 1060 Huayuan Road, Suzhou 215131, Jiangsu, China
| | - Chenhui Zhang
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
- Orthopaedic Institute, Soochow University, 708 Renmin Road, Suzhou 215006, Jiangsu, China
| | - Jun Lin
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
- Department of Orthopaedics, Suzhou Dushu Lake Hospital, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou 215001, Jiangsu, China
| | - Kece Shi
- Department of Orthopaedics, People's Hospital of Leshan, 238 Baita Road, Leshan 614000, Sichuan, China
| | - Zongping Luo
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
- Orthopaedic Institute, Soochow University, 708 Renmin Road, Suzhou 215006, Jiangsu, China
| | - Naicheng Liu
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Xinlin Su
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| |
Collapse
|