151
|
Tumor Cell-Intrinsic Immunometabolism and Precision Nutrition in Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12071757. [PMID: 32630618 PMCID: PMC7409312 DOI: 10.3390/cancers12071757] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022] Open
Abstract
One of the greatest challenges in the cancer immunotherapy field is the need to biologically rationalize and broaden the clinical utility of immune checkpoint inhibitors (ICIs). The balance between metabolism and immune response has critical implications for overcoming the major weaknesses of ICIs, including their lack of universality and durability. The last decade has seen tremendous advances in understanding how the immune system's ability to kill tumor cells requires the conspicuous metabolic specialization of T-cells. We have learned that cancer cell-associated metabolic activities trigger shifts in the abundance of some metabolites with immunosuppressory roles in the tumor microenvironment. Yet very little is known about the tumor cell-intrinsic metabolic traits that control the immune checkpoint contexture in cancer cells. Likewise, we lack a comprehensive understanding of how systemic metabolic perturbations in response to dietary interventions can reprogram the immune checkpoint landscape of tumor cells. We here review state-of-the-art molecular- and functional-level interrogation approaches to uncover how cell-autonomous metabolic traits and diet-mediated changes in nutrient availability and utilization might delineate new cancer cell-intrinsic metabolic dependencies of tumor immunogenicity. We propose that clinical monitoring and in-depth molecular evaluation of the cancer cell-intrinsic metabolic traits involved in primary, adaptive, and acquired resistance to cancer immunotherapy can provide the basis for improvements in therapeutic responses to ICIs. Overall, these approaches might guide the use of metabolic therapeutics and dietary approaches as novel strategies to broaden the spectrum of cancer patients and indications that can be effectively treated with ICI-based cancer immunotherapy.
Collapse
|
152
|
Mangali S, Bhat A, Jadhav K, Kalra J, Sriram D, Vamsi Krishna Venuganti V, Dhar A. Upregulation of PKR pathway mediates glucolipotoxicity induced diabetic cardiomyopathy in vivo in wistar rats and in vitro in cultured cardiomyocytes. Biochem Pharmacol 2020; 177:113948. [DOI: 10.1016/j.bcp.2020.113948] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/01/2020] [Indexed: 12/20/2022]
|
153
|
OGT suppresses S6K1-mediated macrophage inflammation and metabolic disturbance. Proc Natl Acad Sci U S A 2020; 117:16616-16625. [PMID: 32601203 DOI: 10.1073/pnas.1916121117] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Enhanced inflammation is believed to contribute to overnutrition-induced metabolic disturbance. Nutrient flux has also been shown to be essential for immune cell activation. Here, we report an unexpected role of nutrient-sensing O-linked β-N-acetylglucosamine (O-GlcNAc) signaling in suppressing macrophage proinflammatory activation and preventing diet-induced metabolic dysfunction. Overnutrition stimulates an increase in O-GlcNAc signaling in macrophages. O-GlcNAc signaling is down-regulated during macrophage proinflammatory activation. Suppressing O-GlcNAc signaling by O-GlcNAc transferase (OGT) knockout enhances macrophage proinflammatory polarization, promotes adipose tissue inflammation and lipolysis, increases lipid accumulation in peripheral tissues, and exacerbates tissue-specific and whole-body insulin resistance in high-fat-diet-induced obese mice. OGT inhibits macrophage proinflammatory activation by catalyzing ribosomal protein S6 kinase beta-1 (S6K1) O-GlcNAcylation and suppressing S6K1 phosphorylation and mTORC1 signaling. These findings thus identify macrophage O-GlcNAc signaling as a homeostatic mechanism maintaining whole-body metabolism under overnutrition.
Collapse
|
154
|
Sebastian-Valverde M, Pasinetti GM. The NLRP3 Inflammasome as a Critical Actor in the Inflammaging Process. Cells 2020; 9:cells9061552. [PMID: 32604771 PMCID: PMC7348816 DOI: 10.3390/cells9061552] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/12/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
As a consequence of the considerable increase in the human lifespan over the last century, we are experiencing the appearance and impact of new age-related diseases. The causal relationships between aging and an enhanced susceptibility of suffering from a broad spectrum of diseases need to be better understood. However, one specific shared feature seems to be of capital relevance for most of these conditions: the low-grade chronic inflammatory state inherently associated with aging, i.e., inflammaging. Here, we review the molecular and cellular mechanisms that link aging and inflammaging, focusing on the role of the innate immunity and more concretely on the nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, as well as how the chronic activation of this inflammasome has a detrimental effect on different age-related disorders.
Collapse
Affiliation(s)
| | - Giulio M. Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- James J. Peters Veterans Affairs Medical Center, Bronx, New York, NY 10468, USA
- Correspondence: ; Tel.: +1-212-241-1952
| |
Collapse
|
155
|
Kubra KT, Akhter MS, Uddin MA, Barabutis N. Unfolded protein response in cardiovascular disease. Cell Signal 2020; 73:109699. [PMID: 32592779 DOI: 10.1016/j.cellsig.2020.109699] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/20/2020] [Accepted: 06/21/2020] [Indexed: 12/21/2022]
Abstract
The unfolded protein response (UPR) is a highly conserved molecular machinery, which protects the cells against a diverse variety of stimuli. Activation of this element has been associated with both human health and disease. The purpose of the current manuscript is to provide the most updated information on the involvement of UPR towards the improvement; or deterioration of cardiovascular functions. Since UPR is consisted of three distinct elements, namely the activating transcription factor 6, the protein kinase RNA-like endoplasmic reticulum kinase; and the inositol-requiring enzyme-1α, a highly orchestrated manipulation of those molecular branches may provide new therapeutic possibilities against the severe outcomes of cardiovascular disease.
Collapse
Affiliation(s)
- Khadeja-Tul Kubra
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA
| | - Mohammad S Akhter
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA
| | - Mohammad A Uddin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA.
| |
Collapse
|
156
|
Nono Nankam PA, Blüher M, Kehr S, Klöting N, Krohn K, Adams K, Stadler PF, Mendham AE, Goedecke JH. Distinct abdominal and gluteal adipose tissue transcriptome signatures are altered by exercise training in African women with obesity. Sci Rep 2020; 10:10240. [PMID: 32581226 PMCID: PMC7314771 DOI: 10.1038/s41598-020-66868-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/28/2020] [Indexed: 01/09/2023] Open
Abstract
The differential associations of adipose depots with metabolic risk during obesity have been proposed to be controlled by environmental and genetic factors. We evaluated the regional differences in transcriptome signatures between abdominal (aSAT) and gluteal subcutaneous adipose tissue (gSAT) in obese black South African women and tested the hypothesis that 12-week exercise training alters gene expression patterns in a depot-specific manner. Twelve young women performed 12-weeks of supervised aerobic and resistance training. Pre- and post-intervention measurements included peak oxygen consumption (VO2peak), whole-body composition and unbiased gene expression analysis of SAT depots. VO2peak increased, body weight decreased, and body fat distribution improved with exercise training (p < 0.05). The expression of 15 genes, mainly associated with embryonic development, differed between SAT depots at baseline, whereas 318 genes were differentially expressed post-training (p < 0.05). Four developmental genes were differentially expressed between these depots at both time points (HOXA5, DMRT2, DMRT3 and CSN1S1). Exercise training induced changes in the expression of genes associated with immune and inflammatory responses, and lipid metabolism in gSAT, and muscle-associated processes in aSAT. This study showed differences in developmental processes regulating SAT distribution and expandability of distinct depots, and depot-specific adaptation to exercise training in black South African women with obesity.
Collapse
Affiliation(s)
- Pamela A Nono Nankam
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa. .,Department of Endocrinology, Faculty of Medicine, University of Leipzig, Leipzig, Germany.
| | - Matthias Blüher
- Department of Endocrinology, Faculty of Medicine, University of Leipzig, Leipzig, Germany.,Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Stephanie Kehr
- Bioinformatics Group, Department of Computer Science, and Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany
| | - Nora Klöting
- Department of Endocrinology, Faculty of Medicine, University of Leipzig, Leipzig, Germany.,Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Knut Krohn
- Core Unit DNA-Technologies, Medical Faculty, University Leipzig, Leipzig, Germany
| | - Kevin Adams
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Peter F Stadler
- Bioinformatics Group, Department of Computer Science, and Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany
| | - Amy E Mendham
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa.,Non-communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, Cape Town, South Africa
| | - Julia H Goedecke
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa.,Non-communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, Cape Town, South Africa
| |
Collapse
|
157
|
Qureshi R, Picon-Ruiz M, Aurrekoetxea-Rodriguez I, Nunes de Paiva V, D'Amico M, Yoon H, Radhakrishnan R, Morata-Tarifa C, Ince T, Lippman ME, Thaller SR, Rodgers SE, Kesmodel S, Vivanco MDM, Slingerland JM. The Major Pre- and Postmenopausal Estrogens Play Opposing Roles in Obesity-Driven Mammary Inflammation and Breast Cancer Development. Cell Metab 2020; 31:1154-1172.e9. [PMID: 32492394 DOI: 10.1016/j.cmet.2020.05.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 12/26/2019] [Accepted: 05/11/2020] [Indexed: 12/30/2022]
Abstract
Many inflammation-associated diseases, including cancers, increase in women after menopause and with obesity. In contrast to anti-inflammatory actions of 17β-estradiol, we find estrone, which dominates after menopause, is pro-inflammatory. In human mammary adipocytes, cytokine expression increases with obesity, menopause, and cancer. Adipocyte:cancer cell interaction stimulates estrone- and NFκB-dependent pro-inflammatory cytokine upregulation. Estrone- and 17β-estradiol-driven transcriptomes differ. Estrone:ERα stimulates NFκB-mediated cytokine gene induction; 17β-estradiol opposes this. In obese mice, estrone increases and 17β-estradiol relieves inflammation. Estrone drives more rapid ER+ breast cancer growth in vivo. HSD17B14, which converts 17β-estradiol to estrone, associates with poor ER+ breast cancer outcome. Estrone and HSD17B14 upregulate inflammation, ALDH1 activity, and tumorspheres, while 17β-estradiol and HSD17B14 knockdown oppose these. Finally, a high intratumor estrone:17β-estradiol ratio increases tumor-initiating stem cells and ER+ cancer growth in vivo. These findings help explain why postmenopausal ER+ breast cancer increases with obesity, and offer new strategies for prevention and therapy.
Collapse
Affiliation(s)
- Rehana Qureshi
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA.
| | - Manuel Picon-Ruiz
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA; Department of Human Anatomy and Embryology, Faculty of Medicine, Biopathology and Medicine Regenerative Institute (IBIMER), and Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain; Biosanitary Institute of Granada (ibs.GRANADA), Granada, Spain.
| | - Iskander Aurrekoetxea-Rodriguez
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA; Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Vanessa Nunes de Paiva
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Massimo D'Amico
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Hyunho Yoon
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Ramya Radhakrishnan
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Cynthia Morata-Tarifa
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Tan Ince
- Department of Pathology, Weill Cornell Medicine and New York Presbyterian Brooklyn Methodist Hospital, New York, NY, USA
| | - Marc E Lippman
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA; Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Seth R Thaller
- Department of Plastic Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Steven E Rodgers
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA; Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Susan Kesmodel
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA; Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Maria Del Mar Vivanco
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Joyce M Slingerland
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA; Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
158
|
Tian K, Wang X, Wu Y, Wu X, Du G, Liu W, Wu R. Relationship of tumour-associated macrophages with poor prognosis in Wilms' tumour. J Pediatr Urol 2020; 16:376.e1-376.e8. [PMID: 32299765 DOI: 10.1016/j.jpurol.2020.03.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 03/18/2020] [Indexed: 01/26/2023]
Abstract
BACKGROUND Wilms' tumour (WT) is the most common childhood renal tumour. Tumour-associated macrophages (TAMs) are a critical component of tumour microenvironments and contain two main subtypes, classically (M1) or alternatively (M2) activated macrophages. Evidence has revealed TAMs in predicting poor prognosis in some malignant tumours. However, the role of TAMs in WT is still unclear, and the relationship of different types of TAMs with prognosis has not been elucidated. OBJECTIVE The aim of the study was to explore the presence of two types of TAMs in WT and analyse the relationship of TAMs with prognosis. STUDY DESIGN Overall, 61 paediatric patients with WT underwent nephrectomy before any chemotherapy from April 2006 to March 2014. The tumour tissues were analysed by Western blot, immunohistochemistry, and immunofluorescence to explore the distribution of M1 and M2 macrophages in different stages. Kaplan-Meier analysis with regard to the relationship between the presence of TAMs and follow-up information was performed. RESULTS In the 61 patients (44 males and 17 females), there was a median age of 19 months (IQR 13-35.5); 47 patients are still alive, 11 died, 3 were lost to follow-up. According to the National Wilms Tumor Study (NWTS)-5 guidelines, the distribution of tumour stages was as follows: stage I, 27 patients; stage II, 18 patients; and stage III, 16 patients. The Western blot analysis showed that the density of M1 and M2 macrophages in tumour tissues were significantly greater than that in adjacent normal tissues. Immunohistochemistry showed the proportion of patients with positive M1-type macrophages across different stages: stage I, 66.7% (18/27); stage II, 44.4% (8/18); and stage III, 25% (4/16) (p = 0.027). The proportion of patients with positive M2-type macrophages across different stages: stage I, 25.9% (7/27); stage II, 55.6% (10/18); and stage III, 81.3% (13/16) (p = 0.002). Kaplan-Meier analysis suggested that patients with high densities of M2-type macrophages had shorter overall survival time than those with low densities (log-rank test, p = 0.011). DISCUSSION TAMs play a pivotal comments in the tumour microenvironment and tumorigenesis. With the progression of clinical stage, M2 macrophage densities increased greatly, and M1 macrophage density decreased. M2 macrophages represent a poor prognosis and can be utilized as a new indicator in pathological examination. CONCLUSION There is a high density of TAMs in WT, and M2-type macrophage density increases with tumour progression and implies a poor prognosis.
Collapse
Affiliation(s)
- Kaixuan Tian
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Xiaoqing Wang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Yidi Wu
- School of Medicine, Shandong University, Jinan, Shandong, 250021, PR China
| | - Xiangyu Wu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Guoqiang Du
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Wei Liu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China.
| | - Rongde Wu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China.
| |
Collapse
|
159
|
Jafaripour S, Sedighi S, Jokar MH, Aghaei M, Moradzadeh M. Inflammation, diet, and type 2 diabetes: a mini-review. J Immunoassay Immunochem 2020; 41:768-777. [PMID: 32397924 DOI: 10.1080/15321819.2020.1750423] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Inflammation is a common feature of type 2 diabetes (T2D). Inflammatory cytokines increase in patients with type 2 diabetes, metabolic syndrome, and heart disease. Various types of cells can produce inflammatory cytokines and then release them into the bloodstream, where their complex interactions with target tissues raise a tissue-specific immune response. This review focused on C-reactive protein (CRP), tumor necrosis factor (TNF)-α as an inflammatory cytokine, and adiponectin produced by adipose tissues. Despite the major role of cytokines in the development of T2D, further studies are required to investigate the possible effects of the macronutrient composition of diet on these cytokines.
Collapse
Affiliation(s)
- Simin Jafaripour
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences , Mashhad, Iran
| | - Sima Sedighi
- Golestan Rheumatology Research Center, Golestan University of Medical Sciences , Gorgan, Iran
| | - Mohammad Hassan Jokar
- Golestan Rheumatology Research Center, Golestan University of Medical Sciences , Gorgan, Iran
| | - Mehrdad Aghaei
- Golestan Rheumatology Research Center, Golestan University of Medical Sciences , Gorgan, Iran
| | - Maliheh Moradzadeh
- Golestan Rheumatology Research Center, Golestan University of Medical Sciences , Gorgan, Iran
| |
Collapse
|
160
|
Review: Relationships between metabolism and neutrophil function in dairy cows in the peripartum period. Animal 2020; 14:s44-s54. [PMID: 32024567 DOI: 10.1017/s1751731119003227] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Aspects of neutrophil function are diminished or dysregulated in dairy cows in the weeks just before and after calving, which appears to be an important contributor to the occurrence of retained placenta, mastitis, metritis and endometritis. The timing and mechanisms by which specific elements of neutrophil function are impaired are only partially understood. Oxidative burst capacity is the element of neutrophil function most consistently shown to be impaired in the week after calving, but that observation may partially be biased because oxidative burst has been studied more than other functions. There is sufficient evidence to conclude that the availability of calcium and glucose, and exposure to elevated concentrations of non-esterified fatty acids or β-hydroxybutyrate affect some aspects of neutrophil function. However, these factors have mostly been studied in isolation and their effects are not consistent. Social stressors such as a competitive environment for feeding or lying space should plausibly impair innate immune function, but when studied under controlled conditions such effects have generally not been produced. Similarly, treatment with recombinant bovine granulocyte colony-stimulating factor consistently produces large increases in circulating neutrophil count with modest improvements in function, but this does not consistently reduce the incidence of clinical diseases thought to be importantly attributable to impaired innate immunity. Research is now needed that considers the interactions among known and putative risk factors for impaired neutrophil function in dairy cows in the transition period.
Collapse
|
161
|
Urasaki Y, Beaumont C, Talbot JN, Hill DK, Le TT. Akt3 Regulates the Tissue-Specific Response to Copaiba Essential Oil. Int J Mol Sci 2020; 21:ijms21082851. [PMID: 32325885 PMCID: PMC7216139 DOI: 10.3390/ijms21082851] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/17/2020] [Accepted: 04/18/2020] [Indexed: 12/18/2022] Open
Abstract
This study reports a relationship between Akt3 expression and tissue-specific regulation of the pI3K/Akt/mTOR signaling pathway by copaiba essential oil. Akt3, a protein kinase B isoform important for the regulation of neuronal development, exhibited differential expression levels in cells of various origins. In neuronal and microglial cells, where Akt3 is present, copaiba essential oil positively regulated the pI3K/Akt/mTOR signaling pathway. In contrast, in liver cells and T lymphocytes, where Akt3 is absent, copaiba essential oil negatively regulated the pI3K/Akt/mTOR signaling pathway. The expression of Akt3 via plasmid DNA in liver cells led to positive regulatory effects by copaiba essential oil on the pI3K/Akt/mTOR signaling pathway. In contrast, inhibition of Akt3 expression in neuronal cells via small interfering RNA molecules targeting Akt3 transcripts abrogated the regulatory effects of copaiba essential oil on the pI3K/Akt/mTOR signaling pathway. Interestingly, Akt3 expression did not impact the regulatory effects of copaiba essential oil on other signaling pathways. For example, copaiba essential oil consistently upregulated the MAPK and JAK/STAT signaling pathways in all evaluated cell types, independent of the Akt3 expression level. Collectively, the data indicated that Akt3 expression was required for the positive regulatory effects of copaiba essential oil, specifically on the pI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yasuyo Urasaki
- College of Pharmacy, Roseman University of Health Sciences, 10530 Discovery Drive, Las Vegas, NV 89135, USA; (Y.U.); (J.N.T.)
| | - Cody Beaumont
- dōTERRA International, LLC, 389 South 1300 West, Pleasant Grove, UT 84062, USA; (C.B.); (D.K.H.)
| | - Jeffery N. Talbot
- College of Pharmacy, Roseman University of Health Sciences, 10530 Discovery Drive, Las Vegas, NV 89135, USA; (Y.U.); (J.N.T.)
| | - David K. Hill
- dōTERRA International, LLC, 389 South 1300 West, Pleasant Grove, UT 84062, USA; (C.B.); (D.K.H.)
| | - Thuc T. Le
- College of Pharmacy, Roseman University of Health Sciences, 10530 Discovery Drive, Las Vegas, NV 89135, USA; (Y.U.); (J.N.T.)
- Correspondence: ; Tel.: +1-702-802-2820
| |
Collapse
|
162
|
Hou H, Yang W, Bao S, Cao Y. Epigallocatechin Gallate Suppresses Inflammatory Responses by Inhibiting Toll-like Receptor 4 Signaling and Alleviates Insulin Resistance in the Livers of High-fat-diet Rats. J Oleo Sci 2020; 69:479-486. [PMID: 32281563 DOI: 10.5650/jos.ess19303] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
EGCG is a major pharmacological compound in green tea. Nonalcoholic fatty liver disease (NAFLD) is one of the most common liver diseases worldwide. Inflammation and insulin resistance are involved in the development of the disease. In this study, we investigated the beneficial effect of EGCG on the liver tissue of NAFLD rats induced by a high-fat diet and its underlying mechanism. Thirty Sprague-Dawley rats received a normal diet, a HFD and a HFD+EGCG. The expression levels of inflammatory signaling pathway genes (e.g., TLR4, TRAF6, IKKβ, NF-κB, TNF-α) and insulin signaling transduction pathway genes (e.g., PI3K, AKT, IRS-1, IRS-2) were detected in the liver. We observed that EGCG decreased the triglyceride (TG) concentration in rat livers and suppressed TLR4, TRAF6, IKKβ, p-IKKβ, p-NF-κB, and TNF-α levels compared with those in the HFD group, whereas PI3K, AKT, IRS-1, and IRS-2 indicators were improved. EGCG improves obesity-associated subacute hepatic inflammation states, probably through the TLR4 signaling pathway. Furthermore, EGCG also alleviated hepatic insulin resistance. These data indicate that EGCG improves NAFLD from two ways: inhibition of inflammation and improvement of insulin resistance in liver tissues.
Collapse
Affiliation(s)
- Huimin Hou
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University
| | - Wanli Yang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University
| | - Suqing Bao
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University
| | - Yanli Cao
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, China Medical University
| |
Collapse
|
163
|
Ijaz MU, Ahmad MI, Hussain M, Khan IA, Zhao D, Li C. Meat Protein in High-Fat Diet Induces Adipogensis and Dyslipidemia by Altering Gut Microbiota and Endocannabinoid Dysregulation in the Adipose Tissue of Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:3933-3946. [PMID: 32148030 DOI: 10.1021/acs.jafc.0c00017] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Endocannabinoids modulate insulin and adipokine expression in adipocytes through cannabinoid receptors and their levels are elevated during hyperglycemia and obesity, but little is known about how diets affect them. We assessed the effects of dietary casein, chicken, beef, and pork proteins in a high-fat diet mode, on endocannabinoids, adipogenesis, and biomarkers associated with dyslipemdia. A high-fat beef or chicken diet upregulated cannabinoid 1 receptor, N-acyl phosphatidylethanolamine-selective phospholipase-D and diacylglycerol lipase α in adipose tissue and reduced the immunoreactivity of mitochondrial uncoupling protein 1 in brown adipose tissue. In addition, the high-fat diets with beef and chicken protein had a significant impact on adipocyte differentiation and mitochondrial biogenesis in obese mice. A 16S rRNA gene sequencing indicated that high-fat diets, regardless of the protein source, significantly enhanced the ratio of Firmicutes to Bacteroidetes in colon. Meat proteins in a high-fat diet significantly decreased the relative abundances of Akkermansia and Bifidobacteria but enhanced the lipopolysaccharides level in the serum, which promoted the adipogenesis process by causing dysregulation in the endocannabinoid receptors. Consumption of meat protein with high-fat-induced adiposity, visceral obesity, and dyslipidemia reduced the thermogenesis and had a distinctive effect on the mitochondrial biogenesis compared with casein protein.
Collapse
Affiliation(s)
- Muhammad Umair Ijaz
- Key Laboratory of Meat Processing and Quality Control, MOE, Nanjing Agricultural University, 210095 Nanjing, China
- Key Laboratory of Meat Processing, MARA, Nanjing Agricultural University, 210095 Nanjing, China
- Jiangsu Collaborative Innovation Center of Meat Production, Processing and Quality Control, Nanjing Agricultural University, 210095 Nanjing, China
- College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Muhammad Ijaz Ahmad
- Key Laboratory of Meat Processing and Quality Control, MOE, Nanjing Agricultural University, 210095 Nanjing, China
- Key Laboratory of Meat Processing, MARA, Nanjing Agricultural University, 210095 Nanjing, China
- Jiangsu Collaborative Innovation Center of Meat Production, Processing and Quality Control, Nanjing Agricultural University, 210095 Nanjing, China
- College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Muzhair Hussain
- Key Laboratory of Meat Processing and Quality Control, MOE, Nanjing Agricultural University, 210095 Nanjing, China
- Key Laboratory of Meat Processing, MARA, Nanjing Agricultural University, 210095 Nanjing, China
- Jiangsu Collaborative Innovation Center of Meat Production, Processing and Quality Control, Nanjing Agricultural University, 210095 Nanjing, China
- College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Iftikhar Ali Khan
- Key Laboratory of Meat Processing and Quality Control, MOE, Nanjing Agricultural University, 210095 Nanjing, China
- Key Laboratory of Meat Processing, MARA, Nanjing Agricultural University, 210095 Nanjing, China
- Jiangsu Collaborative Innovation Center of Meat Production, Processing and Quality Control, Nanjing Agricultural University, 210095 Nanjing, China
- College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Di Zhao
- Key Laboratory of Meat Processing and Quality Control, MOE, Nanjing Agricultural University, 210095 Nanjing, China
- Key Laboratory of Meat Processing, MARA, Nanjing Agricultural University, 210095 Nanjing, China
- Jiangsu Collaborative Innovation Center of Meat Production, Processing and Quality Control, Nanjing Agricultural University, 210095 Nanjing, China
- College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Chunbao Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Nanjing Agricultural University, 210095 Nanjing, China
- Key Laboratory of Meat Processing, MARA, Nanjing Agricultural University, 210095 Nanjing, China
- Jiangsu Collaborative Innovation Center of Meat Production, Processing and Quality Control, Nanjing Agricultural University, 210095 Nanjing, China
- College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, China
- International Joint Laboratory of Animal Health and Food Safety, MOE, Nanjing Agricultural University, 210095 Nanjing, China
- National Center for International Research on Animal Gut Nutrition, MOST, Nanjing Agricultural University, 210095 Nanjing, China
| |
Collapse
|
164
|
Festuccia WT. Regulation of Adipocyte and Macrophage Functions by mTORC1 and 2 in Metabolic Diseases. Mol Nutr Food Res 2020; 65:e1900768. [DOI: 10.1002/mnfr.201900768] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/06/2020] [Indexed: 12/13/2022]
Affiliation(s)
- William T. Festuccia
- Department of Physiology and Biophysics Institute of Biomedical Sciences University of Sao Paulo Sao Paulo 05508000 Brazil
| |
Collapse
|
165
|
Nutrient mTORC1 signaling contributes to hepatic lipid metabolism in the pathogenesis of non-alcoholic fatty liver disease. LIVER RESEARCH 2020. [DOI: 10.1016/j.livres.2020.02.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
166
|
Li Y, Wang J, Huang C, Shen M, Zhan H, Xu K. RNA N6-methyladenosine: a promising molecular target in metabolic diseases. Cell Biosci 2020; 10:19. [PMID: 32110378 PMCID: PMC7035649 DOI: 10.1186/s13578-020-00385-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/11/2020] [Indexed: 12/12/2022] Open
Abstract
N6-methyladenosine is a prevalent and abundant transcriptome modification, and its methylation regulates the various aspects of RNAs, including transcription, translation, processing and metabolism. The methylation of N6-methyladenosine is highly associated with numerous cellular processes, which plays important roles in the development of physiological process and diseases. The high prevalence of metabolic diseases poses a serious threat to human health, but its pathological mechanisms remain poorly understood. Recent studies have reported that the progression of metabolic diseases is closely related to the expression of RNA N6-methyladenosine modification. In this review, we aim to summarize the biological and clinical significance of RNA N6-methyladenosine modification in metabolic diseases, including obesity, type 2 diabetes, non-alcoholic fatty liver disease, hypertension, cardiovascular diseases, osteoporosis and immune-related metabolic diseases.
Collapse
Affiliation(s)
- Yan Li
- 1Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan China
| | - Jiawen Wang
- 1Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan China
| | - Chunyan Huang
- Houjie Hospital of Dongguan, Dongguan, 523945 Guangdong China
| | - Meng Shen
- Chengdu Tumor Hospital, Chengdu, 610041 Sichuan China
| | - Huakui Zhan
- 1Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072 Sichuan China
| | - Keyang Xu
- 4Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310023 Zhejiang China
| |
Collapse
|
167
|
Antioxidant Effects and Mechanisms of Medicinal Plants and Their Bioactive Compounds for the Prevention and Treatment of Type 2 Diabetes: An Updated Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1356893. [PMID: 32148647 PMCID: PMC7042557 DOI: 10.1155/2020/1356893] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/31/2019] [Accepted: 01/16/2020] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus is a metabolic disorder that majorly affects the endocrine gland, and it is symbolized by hyperglycemia and glucose intolerance owing to deficient insulin secretory responses and beta cell dysfunction. This ailment affects as many as 451 million people worldwide, and it is also one of the leading causes of death. In spite of the immense advances made in the development of orthodox antidiabetic drugs, these drugs are often considered not successful for the management and treatment of T2DM due to the myriad side effects associated with them. Thus, the exploration of medicinal herbs and natural products as therapeutic sources for the treatment of T2DM is promoted because they have little or no side effects. Bioactive molecules isolated from natural sources have been proven to lower blood glucose levels via regulating one or more of the following mechanisms: improvement of beta cell function, insulin resistance, glucose (re)absorption, and glucagon-like peptide-1 homeostasis. In recent times, the mechanisms of action of different bioactive molecules with antidiabetic properties and phytochemistry are gaining a lot of attention in the area of drug discovery. This review article presents an update of the findings from clinical research into medicinal plant therapy for T2DM.
Collapse
|
168
|
The association between sugar-sweetened beverages intake, body mass index, and inflammation in US adults. Int J Public Health 2020; 65:45-53. [PMID: 31982934 DOI: 10.1007/s00038-020-01330-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/07/2019] [Accepted: 01/02/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES This study aims to (1) assess the associations between sugar-sweetened beverages (SSB) consumption and C-reactive protein (CRP) levels, and (2) evaluate the modifying effect of body mass index (BMI) on the association between SSB consumption and CRP levels. METHODS A total of 6856 eligible adults were selected from the 2007-2010 National Health and Nutrition Examination Survey (NHANES). Average quantity of SSB consumption was calculated from 2-day 24-h dietary recalls. All data analyses were performed with appropriate sampling weights. RESULTS Compared with non-SSB drinkers, a 0.26 mg/l higher CRP was observed in heavy SSB drinkers after adjusting for demographic characteristics, lifestyle patterns, and BMI. An effect modification of BMI on SSB intake and CRP levels was detected (P < 0.05). Medium and heavy SSB consumers with obesity had 0.58 and 0.50 higher CRP than non-SSB consumers, respectively (P = 0.014 and 0.013). No association was found in SSB drinkers who were normal weight or overweight. CONCLUSIONS These findings emphasize that SSB intake is positively associated with CRP levels. Obesity might strengthen CRP levels in individuals with medium/heavy amount of SSB consumption.
Collapse
|
169
|
Pascottini OB, LeBlanc SJ. Modulation of immune function in the bovine uterus peripartum. Theriogenology 2020; 150:193-200. [PMID: 31987594 DOI: 10.1016/j.theriogenology.2020.01.042] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 12/30/2022]
Abstract
There is a high risk of clinical or subclinical reproductive tract disease in the postpartum period in dairy cows. An integrated process of adaptive events should occur synchronously, including a robust but well-regulated immune response in the uterus. Failure of this process may result in reproductive tract inflammatory disease. Up to half of postpartum dairy cows are affected by metritis, purulent vaginal discharge (PVD), or subclinical endometritis. After parturition there is damage to the birth canal, the superficial layer of the endometrium is naturally wounded, and essentially all dairy cows have bacterial contamination in the uterus. Neutrophils are the most abundant type of inflammatory cell and the main line of defence against infection in the uterus. A prompt influx of neutrophils is associated with uterine health. Avoidance of clinical disease (metritis and PVD) depends in large part on how effective the immune response is at limiting the burden and effects of bacterial pathogens, while the occurrence of subclinical endometritis is more a function of avoiding excessive or persistent inflammation. Glucose supply, hypocalcemia, lipid mobilization from body fat, ketosis, and the flux of pro-inflammatory cytokines influence immune response and change rapidly and variably among individual cows. Effective but well-regulated inflammatory response will be favoured by best management practices for transition cows, but specific interventions to modulate immune response to prevent uterine disease remain developmental.
Collapse
Affiliation(s)
| | - Stephen J LeBlanc
- Department of Population Medicine, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
170
|
Onat UI, Yildirim AD, Tufanli Ö, Çimen I, Kocatürk B, Veli Z, Hamid SM, Shimada K, Chen S, Sin J, Shah PK, Gottlieb RA, Arditi M, Erbay E. Intercepting the Lipid-Induced Integrated Stress Response Reduces Atherosclerosis. J Am Coll Cardiol 2020; 73:1149-1169. [PMID: 30871699 PMCID: PMC6424590 DOI: 10.1016/j.jacc.2018.12.055] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/14/2022]
Abstract
Background Eukaryotic cells can respond to diverse stimuli by converging at serine-51 phosphorylation on eukaryotic initiation factor 2 alpha (eIF2α) and activate the integrated stress response (ISR). This is a key step in translational control and must be tightly regulated; however, persistent eIF2α phosphorylation is observed in mouse and human atheroma. Objectives Potent ISR inhibitors that modulate neurodegenerative disorders have been identified. Here, the authors evaluated the potential benefits of intercepting ISR in a chronic metabolic and inflammatory disease, atherosclerosis. Methods The authors investigated ISR’s role in lipid-induced inflammasome activation and atherogenesis by taking advantage of 3 different small molecules and the ATP-analog sensitive kinase allele technology to intercept ISR at multiple molecular nodes. Results The results show lipid-activated eIF2α signaling induces a mitochondrial protease, Lon protease 1 (LONP1), that degrades phosphatase and tensin-induced putative kinase 1 and blocks Parkin-mediated mitophagy, resulting in greater mitochondrial oxidative stress, inflammasome activation, and interleukin-1β secretion in macrophages. Furthermore, ISR inhibitors suppress hyperlipidemia-induced inflammasome activation and inflammation, and reduce atherosclerosis. Conclusions These results reveal endoplasmic reticulum controls mitochondrial clearance by activating eIF2α-LONP1 signaling, contributing to an amplified oxidative stress response that triggers robust inflammasome activation and interleukin-1β secretion by dietary fats. These findings underscore the intricate exchange of information and coordination of both organelles’ responses to lipids is important for metabolic health. Modulation of ISR to alleviate organelle stress can prevent inflammasome activation by dietary fats and may be a strategy to reduce lipid-induced inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Umut I Onat
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey; National Nanotechnology Center, Bilkent University, Ankara, Turkey
| | - Asli D Yildirim
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey; National Nanotechnology Center, Bilkent University, Ankara, Turkey; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California; Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Özlem Tufanli
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey; National Nanotechnology Center, Bilkent University, Ankara, Turkey
| | - Ismail Çimen
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey; National Nanotechnology Center, Bilkent University, Ankara, Turkey
| | - Begüm Kocatürk
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey; National Nanotechnology Center, Bilkent University, Ankara, Turkey; Department of Pediatrics, Division of Infectious Diseases and Immunology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Zehra Veli
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey; National Nanotechnology Center, Bilkent University, Ankara, Turkey
| | - Syed M Hamid
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California; Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kenichi Shimada
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California; Division of Cardiology, Oppenheimer Atherosclerosis Research Center and Atherosclerosis Prevention and Treatment Center, Cedars-Sinai Medical Center, Los Angeles, California; Departments of Medicine and Pediatrics, Division of Pediatric Infectious Diseases, Cedars-Sinai Medical Center, Los Angeles, California
| | - Shuang Chen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California; Departments of Medicine and Pediatrics, Division of Pediatric Infectious Diseases, Cedars-Sinai Medical Center, Los Angeles, California; David Geffen School of Medicine, University of California, Los Angeles, California
| | - Jon Sin
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California; Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Prediman K Shah
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California; Division of Cardiology, Oppenheimer Atherosclerosis Research Center and Atherosclerosis Prevention and Treatment Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Roberta A Gottlieb
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California; Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Moshe Arditi
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California; Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California; Departments of Medicine and Pediatrics, Division of Pediatric Infectious Diseases, Cedars-Sinai Medical Center, Los Angeles, California; David Geffen School of Medicine, University of California, Los Angeles, California
| | - Ebru Erbay
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey; National Nanotechnology Center, Bilkent University, Ankara, Turkey; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California; Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
171
|
Pearson RC, Olenick AA, Green ES, Jenkins NT. Tabata-style functional exercise increases resting and postprandial fat oxidation but does not reduce triglyceride concentrations. Exp Physiol 2020; 105:468-476. [PMID: 31916294 DOI: 10.1113/ep088330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/06/2020] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? What are the metabolic impacts of high intensity functional Tabata exercise? What is the main finding and its importance? Tabata exercise with high intensity functional movements causes increases in fasted and postprandial fat oxidation the day after exercise without altering postprandial triglyceride concentrations. These results support the usage of a Tabata-style high intensity functional exercise to improve postprandial fat oxidation. ABSTRACT We evaluated the effect of a high fat meal with and without prior high intensity functional exercise executed in a Tabata-style interval pattern on resting and postprandial substrate oxidation, as well as postprandial blood glucose and triglyceride concentrations. Eleven healthy males completed two trials (Tabata exercise (TE) and non-exercise control (CON)) in random order separated by 7 days. A two-day protocol was used in which TE or CON was performed on the first day and a high fat meal was administered ∼13 h later the following morning. Power output from the TE session was quantified using a kinematic approach by calculating external work performed per unit time for each of the four exercises (rowing, dumbbell thrusters, kettlebell swings and burpees). For the meal challenge, respiratory gases and blood samples were taken fasted and at 1, 2 and 3 h postprandial. Fat oxidation was significantly higher after TE compared to CON at all time points (P < 0.05). Carbohydrate oxidation was significantly lower after TE compared to CON at 1 h postprandial (P < 0.05). There were no significant effects of TE on fasting or postprandial glucose or triglyceride concentrations. Functional exercises performed in a high intensity TE pattern enhance fasting and postprandial fat oxidation on the following day with minimal influence on blood triglycerides or glucose levels.
Collapse
Affiliation(s)
- Regis C Pearson
- Department of Kinesiology, College of Education, University of Georgia, 330 River Rd, Athens, GA, 30602, USA
| | - Alyssa A Olenick
- Department of Kinesiology, College of Education, University of Georgia, 330 River Rd, Athens, GA, 30602, USA
| | - Edward S Green
- Department of Kinesiology, College of Education, University of Georgia, 330 River Rd, Athens, GA, 30602, USA
| | - Nathan T Jenkins
- Department of Kinesiology, College of Education, University of Georgia, 330 River Rd, Athens, GA, 30602, USA
| |
Collapse
|
172
|
Martins VD, Silva FC, Caixeta F, Carneiro MB, Goes GR, Torres L, Barbosa SC, Vaz L, Paiva NC, Carneiro CM, Vieira LQ, Faria AMC, Maioli TU. Obesity impairs resistance to Leishmania major infection in C57BL/6 mice. PLoS Negl Trop Dis 2020; 14:e0006596. [PMID: 31923234 PMCID: PMC6953764 DOI: 10.1371/journal.pntd.0006596] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/28/2019] [Indexed: 01/19/2023] Open
Abstract
An association between increased susceptibility to infectious diseases and obesity has been described as a result of impaired immunity in obese individuals. It is not clear whether a similar linkage can be drawn between obesity and parasitic diseases. To evaluate the effect of obesity in the immune response to cutaneous Leishmania major infection, we studied the ability of C57BL/6 mice fed a hypercaloric diet (HSB) to control leishmaniasis. Mice with diet-induced obesity presented thicker lesions with higher parasite burden and a more intense inflammatory infiltrate in the infected ear after infection with L. major. There was no difference between control and obese mice in IFN-gamma or IL-4 production by auricular draining lymph node cells, but obese mice produced higher levels of IgG1 and IL-17. Peritoneal macrophages from obese mice were less efficient to kill L. major when infected in vitro than macrophages from control mice. In vitro stimulation of macrophages with IL-17 decreased their capacity to kill the parasite. Moreover, macrophages from obese mice presented higher arginase activity. To confirm the role of IL-17 in the context of obesity and infection, we studied lesion development in obese IL-17R-/- mice infected with L. major and found no difference in skin lesions and the leukocyte accumulation in the draining lymph node is redcuced in knockout mice compared between obese and lean animals. Our results indicate that diet-induced obesity impairs resistance to L. major in C57BL/6 mice and that IL-17 is involved in lesion development. Obesity is a serious and increasing public health problem, and also induces a spectrum of metabolic disorders. Some diseases are known to be more severe in the presence of obesity. However, the interactions of obesity with the immune response to infectious agents have not been fully explored. In this study, we investigated the response of obese mice to infection with Leishmania major. C57BL/6 mice were fed a hypercaloric diet (HSB) and infected afterward with L. major. In obese mice, lesions were ticker and more ulcerative, and cells from draining lymph nodes produced more IL-17 when compared with cells from lean mice fed a control diet. Macrophages from obese and lean mice were infected in vitro and stimulated with IL-17 to test the role of this cytokine in effect produced by obesity. Macrophages from obese mice were more infected by L. major than the macrophages from control mice and the number of parasites was increased by treatment with IL-17. IL-17R deficient mice treated with hypercaloric diet showed no difference in lesion size when compared to mice fed control diet. Our findings suggest that diet-induced obesity decrease the resistance to L. major infection of C57BL/6 mice and the IL-17 cytokine may be involved in the lesion formation.
Collapse
Affiliation(s)
- Vinicius Dantas Martins
- Programa de Pós-Graduação em Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Franciele Carolina Silva
- Programa de Pós-Graduação em Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Felipe Caixeta
- Programa de Pós-Graduação Interunidades de Bioinformática, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Matheus Batista Carneiro
- Programa de Pós-Graduação em Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Disease, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Graziele Ribeiro Goes
- Programa de Pós-Graduação em Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lícia Torres
- Programa de Pós-Graduação em Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sara Cândida Barbosa
- Programa de Pós-Graduação em Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo Vaz
- Programa de Pós-Graduação em Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Nivea Carolina Paiva
- Núcleo de Pesquisa em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Cláudia Martins Carneiro
- Núcleo de Pesquisa em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Leda Quercia Vieira
- Programa de Pós-Graduação em Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Maria Caetano Faria
- Programa de Pós-Graduação em Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tatiani Uceli Maioli
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- * E-mail:
| |
Collapse
|
173
|
Martínez-García MÁ, Ojeda-Ojeda M, Rodríguez-Martín E, Insenser M, Moncayo S, Álvarez-Blasco F, Luque-Ramírez M, Escobar-Morreale HF. TLR2 and TLR4 Surface and Gene Expression in White Blood Cells after Fasting and Oral Glucose, Lipid and Protein Challenges: Influence of Obesity and Sex Hormones. Biomolecules 2020; 10:biom10010111. [PMID: 31936430 PMCID: PMC7023426 DOI: 10.3390/biom10010111] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 12/20/2019] [Accepted: 01/04/2020] [Indexed: 12/19/2022] Open
Abstract
We studied if macronutrients of the diet have different effects on leukocyte activation, and if these effects are influenced by sex hormones or obesity. We analyzed leukocyte cell surface and gene expression of toll-like receptors 2 and 4 (TLR2 and TLR4) during fasting and after macronutrient loads in women with polycystic ovary syndrome and female and male controls. Fasting TLR2 surface expression in neutrophils was higher in men than in women. Obese subjects presented higher TLR2 gene expression than nonobese individuals, particularly in men. In contrast, surface TLR4 expression was lower in men and in obese individuals. Postprandial cell-surface expression decreased similarly after all macronutrient loads. Neutrophil TLR2 decreased only in obese subjects whereas TLR4 showed a greater decrease in nonobese individuals. However, TLR2 gene expression increased after glucose ingestion and decreased during the lipid load, while TLR4 was induced in response to lipids and mostly to glucose. Postprandial TLR gene expression was not influenced by group of subjects or obesity. Both cell-surface and gene postprandial expression inversely correlated with their fasting levels. These responses suggest a transient compensatory response aiming to prevent postprandial inflammation. However, obesity and sex hormones showed opposite influences on surface expression of TLR2 and TLR4, but not on their gene expression, pointing to regulatory posttranscriptional mechanisms.
Collapse
Affiliation(s)
- M. Ángeles Martínez-García
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
| | - Miriam Ojeda-Ojeda
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
| | | | - María Insenser
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
| | - Samuel Moncayo
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
| | - Francisco Álvarez-Blasco
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
| | - Manuel Luque-Ramírez
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
| | - Héctor F. Escobar-Morreale
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal &Universidad de Alcalá &Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28034 Madrid, Spain; (M.Á.M.-G.); (M.O.-O.); (M.I.); (S.M.); (F.Á.-B.); (M.L.-R.)
- Correspondence: ; Tel.: +34-91-3369164
| |
Collapse
|
174
|
Rocha-Ramírez LM, Hernández-Ochoa B, Gómez-Manzo S, Marcial-Quino J, Cárdenas-Rodríguez N, Centeno-Leija S, García-Garibay M. Evaluation of Immunomodulatory Activities of the Heat-Killed Probiotic Strain Lactobacillus casei IMAU60214 on Macrophages In Vitro. Microorganisms 2020; 8:microorganisms8010079. [PMID: 31936101 PMCID: PMC7022880 DOI: 10.3390/microorganisms8010079] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/02/2020] [Accepted: 01/05/2020] [Indexed: 12/16/2022] Open
Abstract
Most Lactobacillus species have beneficial immunological (“immunoprobiotic”) effects in the host. However, it is unclear how probiotic bacteria regulate immune responses. The present study investigated the effects of heat-killed Lactobacillus casei IMAU60214 on the activity of human monocyte-derived macrophages (MDMs). Human MDMs were treated with heat-killed L. casei at a ratio (bacteria/MDM) of 50:1, 100:1, 250:1, and 500:1, and then evaluated for the following: NO production, by Griess reaction; phagocytosis of FITC-labeled Staphylococcus aureus particles; cytokine secretion profile (tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-12p70, IL-10, and transforming growth factor (TGF)-β) by ELISA; and costimulatory molecule (CD80 and CD86) surface expression, by flow cytometry. Heat-killed L. casei IMAU60214 enhanced phagocytosis, NO production, cytokine release, and surface expression of CD80 and CD86 in a dose-dependent manner. All products were previously suppressed by pretreatment with a Toll-like receptor 2 (TLR2)-neutralizing antibody. Overall, our findings suggest that this probiotic strain promotes an M1-like pro-inflammatory phenotype through the TLR2 signaling pathway. These effects on macrophage phenotype help explain the probiotic efficacy of Lactobacillus and provide important information for the selection of therapeutic targets and treatments compatible with the immunological characteristics of this probiotic strain.
Collapse
Affiliation(s)
- Luz María Rocha-Ramírez
- Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez, Secretaría de Salud Dr. Márquez No. 162, Col Doctores, Delegación Cuauhtémoc, Ciudad de México 06720, Mexico
- Correspondence: ; Tel.: +52-55-5228-9917 (ext. 2084)
| | - Beatriz Hernández-Ochoa
- Laboratorio de Inmunoquímica y Biología Celular, Hospital Infantil de México Federico Gómez, Secretaría de Salud. Dr. Márquez No. 162, Col Doctores, Delegación Cuauhtémoc, Ciudad de México 06720, Mexico;
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaria de Salud, Ciudad de México 04530, Mexico;
| | - Jaime Marcial-Quino
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico;
| | - Noemí Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico;
| | - Sara Centeno-Leija
- Consejo Nacional Ciencia y Tecnologia (CONACYT) Laboratorio de Agrobiotecnología, Tecnoparque CLQ, Universidad de Colima, Carretera Los Limones-Loma de Juárez, Colima 28629, Mexico;
| | - Mariano García-Garibay
- Departamento de Ciencias de la Alimentación, Unidad Lerma, Universidad Autónoma Metropolitana, Av. San Rafael Atlixco No. 186. Col Vicentina, Ciudad de México 09340, Mexico;
| |
Collapse
|
175
|
Harakeh S, Kalamegam G, Pushparaj PN, Al-Hejin A, Alfadul SM, Al Amri T, Barnawi S, Al Sadoun H, Mirza AA, Azhar E. Chemokines and their association with body mass index among healthy Saudis. Saudi J Biol Sci 2020; 27:6-11. [PMID: 31889810 PMCID: PMC6933256 DOI: 10.1016/j.sjbs.2019.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/17/2019] [Accepted: 03/19/2019] [Indexed: 12/23/2022] Open
Abstract
Obesity is a chronic disorder that is associated with body mass index (BMI) of greater or equal to 30 kg/m2. The prevalence of obesity in the Kingdom of Saudi Arabia (KSA) is increasing at an alarming rate and is expected to reach 41% in men and up to 78% in women by 2022. Since chemokines are associated with involuntary weight loss, the objective of this study was to elucidate their association with BMI among Saudis. A questionnaire was used to collect information about diet, health conditions, and demographics from 15 men and 16 women who participated in the study. BMI was calculated based on clinical measurements and participants were classified according to their BMI category as: normal, underweight, overweight, or obese. Serum samples were collected for a multiplex assay using the Human Chemokine Magnetic 30-plex panel. The serum concentration of either the monokine induced by gamma interferon (MIG) or the CXC-motif chemokine ligand 9 (CXCL-9) was significantly increased in obese men (P = 0.0194) and women (P = 0.043) as compared to underweight men and women, respectively. However, the serum levels of other chemokines were not significantly different among the groups. We found that MIG levels are differentially regulated in serum, based on individuals' BMI.
Collapse
Affiliation(s)
- Steve Harakeh
- Special Infectious Agents Unit, King Fahd Medical Research Center, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Gauthaman Kalamegam
- Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Peter N. Pushparaj
- Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ahmed Al-Hejin
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sulaiman M. Alfadul
- King Abdulaziz City for Science and Technology, P.O Box 6086, Riyadh 11442, Saudi Arabia
| | - Turki Al Amri
- Family and Community Medicine Department, Faculty of Medicine-Rabigh Branch, KAU, Jeddah 21589, Saudi Arabia
| | - Salah Barnawi
- Special Infectious Agents Unit, King Fahd Medical Research Center, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hadeel Al Sadoun
- Department of Laboratory Medical Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Saudi Arabia
| | - Ahmed A. Mirza
- Department of Laboratory Medical Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Saudi Arabia
| | - Esam Azhar
- Special Infectious Agents Unit, King Fahd Medical Research Center, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Laboratory Medical Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Saudi Arabia
| |
Collapse
|
176
|
Poon K. Behavioral Feeding Circuit: Dietary Fat-Induced Effects of Inflammatory Mediators in the Hypothalamus. Front Endocrinol (Lausanne) 2020; 11:591559. [PMID: 33324346 PMCID: PMC7726204 DOI: 10.3389/fendo.2020.591559] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/23/2020] [Indexed: 12/19/2022] Open
Abstract
Excessive dietary fat intake has extensive impacts on several physiological systems and can lead to metabolic and nonmetabolic disease. In animal models of ingestion, exposure to a high fat diet during pregnancy predisposes offspring to increase intake of dietary fat and causes increase in weight gain that can lead to obesity, and without intervention, these physiological and behavioral consequences can persist for several generations. The hypothalamus is a region of the brain that responds to physiological hunger and fullness and contains orexigenic neuropeptide systems that have long been associated with dietary fat intake. The past fifteen years of research show that prenatal exposure to a high fat diet increases neurogenesis of these neuropeptide systems in offspring brain and are correlated to behavioral changes that induce a pro-consummatory and obesogenic phenotype. Current research has uncovered several potential molecular mechanisms by which excessive dietary fat alters the hypothalamus and involve dietary fatty acids, the immune system, gut microbiota, and transcriptional and epigenetic changes. This review will examine the current knowledge of dietary fat-associated changes in the hypothalamus and the potential pathways involved in modifying the development of orexigenic peptide neurons that lead to changes in ingestive behavior, with a special emphasis on inflammation by chemokines.
Collapse
|
177
|
Romero-Zerbo SY, García-Fernández M, Espinosa-Jiménez V, Pozo-Morales M, Escamilla-Sánchez A, Sánchez-Salido L, Lara E, Cobo-Vuilleumier N, Rafacho A, Olveira G, Rojo-Martínez G, Gauthier BR, González-Mariscal I, Bermúdez-Silva FJ. The Atypical Cannabinoid Abn-CBD Reduces Inflammation and Protects Liver, Pancreas, and Adipose Tissue in a Mouse Model of Prediabetes and Non-alcoholic Fatty Liver Disease. Front Endocrinol (Lausanne) 2020; 11:103. [PMID: 32210914 PMCID: PMC7067697 DOI: 10.3389/fendo.2020.00103] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/18/2020] [Indexed: 01/07/2023] Open
Abstract
Background and Aims: The synthetic atypical cannabinoid Abn-CBD, a cannabidiol (CBD) derivative, has been recently shown to modulate the immune system in different organs, but its impact in obesity-related meta-inflammation remains unstudied. We investigated the effects of Abn-CBD on metabolic and inflammatory parameters utilizing a diet-induced obese (DIO) mouse model of prediabetes and non-alcoholic fatty liver disease (NAFLD). Materials and Methods: Ten-week-old C57Bl/6J mice were fed a high-fat diet for 15 weeks, following a 2-week treatment of daily intraperitoneal injections with Abn-CBD or vehicle. At week 15 mice were obese, prediabetic and developed NAFLD. Body weight and glucose homeostasis were monitored. Mice were euthanized and blood, liver, adipose tissue and pancreas were collected and processed for metabolic and inflammatory analysis. Results: Body weight and triglycerides profiles in blood and liver were comparable between vehicle- and Abn-CBD-treated DIO mice. However, treatment with Abn-CBD reduced hyperinsulinemia and markers of systemic low-grade inflammation in plasma and fat, also promoting white adipose tissue browning. Pancreatic islets from Abn-CBD-treated mice showed lower apoptosis, inflammation and oxidative stress than vehicle-treated DIO mice, and beta cell proliferation was induced. Furthermore, Abn-CBD lowered hepatic fibrosis, inflammation and macrophage infiltration in the liver when compared to vehicle-treated DIO mice. Importantly, the balance between hepatocyte proliferation and apoptosis was improved in Abn-CBD-treated compared to vehicle-treated DIO mice. Conclusions: These results suggest that Abn-CBD exerts beneficial immunomodulatory actions in the liver, pancreas and adipose tissue of DIO prediabetic mice with NAFLD, thus protecting tissues. Therefore, Abn-CBD and related compounds could represent novel pharmacological strategies for managing obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Silvana Y. Romero-Zerbo
- UGC Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional de Málaga, Universidad de Málaga, Málaga, Spain
| | - María García-Fernández
- Departamento de Fisiología Humana, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga-IBIMA, Universidad de Málaga, Málaga, Spain
| | - Vanesa Espinosa-Jiménez
- UGC Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional de Málaga, Universidad de Málaga, Málaga, Spain
| | - Macarena Pozo-Morales
- UGC Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional de Málaga, Universidad de Málaga, Málaga, Spain
| | | | - Lourdes Sánchez-Salido
- UGC Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional de Málaga, Universidad de Málaga, Málaga, Spain
| | - Estrella Lara
- Departamento de Fisiología Humana, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga-IBIMA, Universidad de Málaga, Málaga, Spain
| | - Nadia Cobo-Vuilleumier
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville, Spain
| | - Alex Rafacho
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, Brazil
| | - Gabriel Olveira
- UGC Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional de Málaga, Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Gemma Rojo-Martínez
- UGC Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional de Málaga, Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Benoit R. Gauthier
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Isabel González-Mariscal
- UGC Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional de Málaga, Universidad de Málaga, Málaga, Spain
- *Correspondence: Isabel González-Mariscal
| | - Francisco J. Bermúdez-Silva
- UGC Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional de Málaga, Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Francisco J. Bermúdez-Silva
| |
Collapse
|
178
|
Novel structured diacylglycerol (DAG) rich oleo formulations activate the Nrf2 pathway and impedes NF-κB translocation to mitigate pre-clinical conditions of hypertension. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
179
|
Wang Z, Ni X, Zhang L, Sun L, Zhu X, Zhou Q, Yang Z, Yuan H. Toll-Like Receptor 4 and Inflammatory Micro-Environment of Pancreatic Islets in Type-2 Diabetes Mellitus: A Therapeutic Perspective. Diabetes Metab Syndr Obes 2020; 13:4261-4272. [PMID: 33204132 PMCID: PMC7666984 DOI: 10.2147/dmso.s279104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Patients with type-2 diabetes mellitus (T2DM) display chronic low-grade inflammation induced by activation of the innate immune system. Toll-like receptor (TLR)4 is a pattern recognition receptor that plays a vital part in activation of the innate immune system. Results from animal and computer-simulation studies have demonstrated that targeting TLR4 to block the TLR4-nuclear factor-kappa B (NF-κB) pathway reduces the inflammatory response and complications associated with T2DM. Therefore, TLR4-targeted therapy has broad prospects. Here, we reviewed the role of TLR4 in inflammation during chronic hyperglycemia in T2DM and its therapeutic prospects.
Collapse
Affiliation(s)
- Zhaoping Wang
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, People’s Republic of China
| | - Xiaolin Ni
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Li Zhang
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, People’s Republic of China
| | - Liang Sun
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, People’s Republic of China
| | - Xiaoquan Zhu
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, People’s Republic of China
| | - Qi Zhou
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, People’s Republic of China
| | - Ze Yang
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, People’s Republic of China
| | - Huiping Yuan
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, People’s Republic of China
- Correspondence: Huiping Yuan The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Dongdan DaHua Road 1#, Beijing100730, People’s Republic of ChinaTel +86-10-58115043Fax +86-10-65237929 Email
| |
Collapse
|
180
|
Laviada-Molina HA, Leal-Berumen I, Rodriguez-Ayala E, Bastarrachea RA. Working Hypothesis for Glucose Metabolism and SARS-CoV-2 Replication: Interplay Between the Hexosamine Pathway and Interferon RF5 Triggering Hyperinflammation. Role of BCG Vaccine? Front Endocrinol (Lausanne) 2020; 11:514. [PMID: 32733388 PMCID: PMC7358362 DOI: 10.3389/fendo.2020.00514] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 06/26/2020] [Indexed: 12/20/2022] Open
Affiliation(s)
| | - Irene Leal-Berumen
- Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Chihuahua, México
| | - Ernesto Rodriguez-Ayala
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, Naucalpan de Juárez, México
| | - Raul A. Bastarrachea
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, United States
- *Correspondence: Raul A. Bastarrachea
| |
Collapse
|
181
|
Herremans KM, Riner AN, Cameron ME, Trevino JG. The Microbiota and Cancer Cachexia. Int J Mol Sci 2019; 20:ijms20246267. [PMID: 31842339 PMCID: PMC6940781 DOI: 10.3390/ijms20246267] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 12/05/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer cachexia is a multifactorial syndrome defined by weight loss, muscle wasting, and systemic inflammation. It affects the majority of patients with advanced cancer and is associated with poor treatment response, early mortality and decreased quality of life. The microbiota has been implicated in cancer cachexia through pathways of systemic inflammation, gut barrier dysfunction and muscle wasting. The imbalance of the microbiota, known as dysbiosis, has been shown to influence cancer cachexia. Bacteria that play beneficial and detrimental roles in the disease pathogenesis have been identified. The phenotype of cancer cachexia is associated with decreased levels of Lactobacillales and increased levels of Enterobacteriaceae and Parabacteroides. Currently, there are no treatment options that demonstrate increased survival or the quality of life in patients suffering from cancer cachexia. Through the manipulation of beneficial bacteria in the gut microbiota, different treatment options have been explored. Prebiotics and probiotics have been shown to improve outcomes in animal models of cachexia. Expounding on this mechanism, fecal microbiota transplant (FMT) holds promise for a future treatment of cancer cachexia. Further research is necessary to address this detrimental disease process and improve the lives of patients suffering from cancer cachexia.
Collapse
|
182
|
Wei CW, Lee CY, Lee DJ, Chu CF, Wang JC, Wang TC, Jane WN, Chang ZF, Leu CM, Dzhagalov IL, Hsu CL. Equilibrative Nucleoside Transporter 3 Regulates T Cell Homeostasis by Coordinating Lysosomal Function with Nucleoside Availability. Cell Rep 2019; 23:2330-2341. [PMID: 29791845 DOI: 10.1016/j.celrep.2018.04.077] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 01/29/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
T cells are a versatile immune cell population responding to challenges by differentiation and proliferation followed by contraction and memory formation. Dynamic metabolic reprogramming is essential for T cells to meet the biosynthetic needs and the reutilization of biomolecules, processes that require active participation of metabolite transporters. Here, we show that equilibrative nucleoside transporter 3 (ENT3) is highly expressed in peripheral T cells and has a key role in maintaining T cell homeostasis by supporting the proliferation and survival of T cells. ENT3 deficiency leads to an enlarged and disturbed lysosomal compartment, resulting in accumulation of surplus mitochondria, elevation of intracellular reactive oxygen species, and DNA damage in T cells. Our results identify ENT3 as a vital metabolite transporter that supports T cell homeostasis and activation by regulating lysosomal integrity and the availability of nucleosides. Moreover, we uncovered that T cell lysosomes are an important source of salvaged metabolites for survival and proliferation.
Collapse
Affiliation(s)
- Chin-Wen Wei
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Chia-Ying Lee
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Ding-Jin Lee
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Chang-Feng Chu
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Ju-Chu Wang
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Tien-Chiao Wang
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Wann-Neng Jane
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 115, Taiwan
| | - Zee-Fen Chang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan; Institute of Molecular Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chuen-Miin Leu
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Ivan L Dzhagalov
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Chia-Lin Hsu
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan.
| |
Collapse
|
183
|
Bouagnon AD, Lin L, Srivastava S, Liu CC, Panda O, Schroeder FC, Srinivasan S, Ashrafi K. Intestinal peroxisomal fatty acid β-oxidation regulates neural serotonin signaling through a feedback mechanism. PLoS Biol 2019; 17:e3000242. [PMID: 31805041 PMCID: PMC6917301 DOI: 10.1371/journal.pbio.3000242] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 12/17/2019] [Accepted: 11/15/2019] [Indexed: 02/02/2023] Open
Abstract
The ability to coordinate behavioral responses with metabolic status is fundamental to the maintenance of energy homeostasis. In numerous species including Caenorhabditis elegans and mammals, neural serotonin signaling regulates a range of food-related behaviors. However, the mechanisms that integrate metabolic information with serotonergic circuits are poorly characterized. Here, we identify metabolic, molecular, and cellular components of a circuit that links peripheral metabolic state to serotonin-regulated behaviors in C. elegans. We find that blocking the entry of fatty acyl coenzyme As (CoAs) into peroxisomal β-oxidation in the intestine blunts the effects of neural serotonin signaling on feeding and egg-laying behaviors. Comparative genomics and metabolomics revealed that interfering with intestinal peroxisomal β-oxidation results in a modest global transcriptional change but significant changes to the metabolome, including a large number of changes in ascaroside and phospholipid species, some of which affect feeding behavior. We also identify body cavity neurons and an ether-a-go-go (EAG)-related potassium channel that functions in these neurons as key cellular components of the circuitry linking peripheral metabolic signals to regulation of neural serotonin signaling. These data raise the possibility that the effects of serotonin on satiety may have their origins in feedback, homeostatic metabolic responses from the periphery.
Collapse
Affiliation(s)
- Aude D. Bouagnon
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
| | - Lin Lin
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
| | - Shubhi Srivastava
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, United States of America
| | - Chung-Chih Liu
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, United States of America
| | - Oishika Panda
- Boyce Thompson Institute, Cornell University, Ithaca, New York, United States of America
| | - Frank C. Schroeder
- Boyce Thompson Institute, Cornell University, Ithaca, New York, United States of America
| | - Supriya Srinivasan
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, United States of America
| | - Kaveh Ashrafi
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
184
|
Ahmed OT, Allen AM. Extrahepatic Malignancies in Nonalcoholic Fatty Liver Disease. CURRENT HEPATOLOGY REPORTS 2019; 18:455-472. [PMID: 36397965 PMCID: PMC9668075 DOI: 10.1007/s11901-019-00499-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
PURPOSE OF REVIEW Malignancy is the second most common cause of death in individuals with nonalcoholic fatty liver disease (NAFLD). Understanding unique characteristics of malignancy risk beyond hepatocellular carcinoma in NAFLD has significant implications in counseling and personalized preventative measures in this high-risk population. Herein, we systematically review the literature reporting extra-hepatic malignancies in NAFLD and discuss the key biological mechanisms underpinning the association between excess adiposity and cancer risk. RECENT FINDINGS Several studies have shown significant associations between NAFLD and extrahepatic malignancies. The strongest association was found with cancers of the gastrointestinal tract and hormone-sensitive cancers. Recent data support sex-specific differences in cancer risk increase in NAFLD: colorectal cancer in men and uterine cancer in women. The risk of cancer development is higher in NAFLD than obesity alone. SUMMARY A growing body of observational evidence over the last decade supports the association between NAFLD and extrahepatic malignancies. This association requires further studies, ideally designed to include more detailed measures of body fat deposition beyond BMI in well-characterized, large cohorts of NAFLD patients, to determine if screening policies should be individualized in this group.
Collapse
Affiliation(s)
- Omar T. Ahmed
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Alina M. Allen
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
185
|
Afroz S, Shama, Battu S, Matin S, Solouki S, Elmore JP, Minhas G, Huang W, August A, Khan N. Amino acid starvation enhances vaccine efficacy by augmenting neutralizing antibody production. Sci Signal 2019; 12:12/607/eaav4717. [PMID: 31719173 DOI: 10.1126/scisignal.aav4717] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Specific reduction in the intake of proteins or amino acids (AAs) offers enormous health benefits, including increased life span, protection against age-associated disorders, and improved metabolic fitness and immunity. Cells respond to conditions of AA starvation by activating the amino acid starvation response (AAR). Here, we showed that mimicking AAR with halofuginone (HF) enhanced the magnitude and affinity of neutralizing, antigen-specific antibody responses in mice immunized with dengue virus envelope domain III protein (DENVrEDIII), a potent vaccine candidate against DENV. HF enhanced the formation of germinal centers (GCs) and increased the production of the cytokine IL-10 in the secondary lymphoid organs of vaccinated mice. Furthermore, HF promoted the transcription of genes associated with memory B cell formation and maintenance and maturation of GCs in the draining lymph nodes of vaccinated mice. The increased abundance of IL-10 in HF-preconditioned mice correlated with enhanced GC responses and may promote the establishment of long-lived plasma cells that secrete antigen-specific, high-affinity antibodies. Thus, these data suggest that mimetics of AA starvation could provide an alternative strategy to augment the efficacy of vaccines against dengue and other infectious diseases.
Collapse
Affiliation(s)
- Sumbul Afroz
- School of Life Sciences, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, 500046 Telangana, India
| | - Shama
- School of Life Sciences, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, 500046 Telangana, India
| | - Srikanth Battu
- School of Life Sciences, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, 500046 Telangana, India
| | - Shaikh Matin
- School of Life Sciences, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, 500046 Telangana, India
| | - Sabrina Solouki
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Jessica P Elmore
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Gillipsie Minhas
- School of Life Sciences, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, 500046 Telangana, India
| | - Weishan Huang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.,Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Nooruddin Khan
- School of Life Sciences, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, 500046 Telangana, India.
| |
Collapse
|
186
|
Innate sensitivity to stress facilitates inflammation, alters metabolism and shortens lifespan in a mouse model of social hierarchy. Aging (Albany NY) 2019; 11:9901-9911. [PMID: 31707362 PMCID: PMC6874436 DOI: 10.18632/aging.102440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/29/2019] [Indexed: 02/07/2023]
Abstract
It is known that stress alters homeostasis and may lead to accelerated aging. However, little is known about the contribution of innate susceptibility to stress to the deterioration of physiological functions, acceleration of aging and developing of age-related diseases. By using socially-submissive stress susceptible (Sub) and socially-dominant stress resilient (Dom) selectively bred mouse model we observed a marked reduction in the lifespan of both male and female Sub mice. We found that innate susceptibility to stress correlates with chronic inflammation, development of splenomegaly and a significant increase in the levels of circulating pro-inflammatory cytokines IL-1β and IL-6. Furthermore, Sub mice showed a marked hypoglycemia, reduction of insulin levels, increase in GSK3 activity and elevation of IGF-1 serum levels, as well as low skin surface temperature and body weight. Interestingly, lifelong exposure of Sub mice to chronic mild stress did not further reduce their lifespan, indicating a high level of intrinsic stress. Taken together, our data reveal that social submissiveness coupled with innate stress sensitivity coincides with inflammation, leading to the deterioration of physiological functions and early aging independent of whether an individual is exposed to stress or not.
Collapse
|
187
|
Shini S, Shini A, Bryden WL. Unravelling fatty liver haemorrhagic syndrome: 2. Inflammation and pathophysiology. Avian Pathol 2019; 49:131-143. [PMID: 31621393 DOI: 10.1080/03079457.2019.1682119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
To study the role of inflammation in the pathophysiology of the fatty liver haemorrhagic syndrome (FLHS), mature laying hens were treated with oestrogen (β-oestradiol-17-dipropionate or E2) and challenged with lipopolysaccharide (LPS). Oestrogen injections induced FLHS, but the incidence and severity of the condition was increased with a combination of E2 & LPS. Hepatic mRNA levels of the genes encoding key regulators of inflammation, such as interleukin-1β (IL-1β), interleukin-6 (IL-6) and interleukin-18 (IL-18), were evaluated. The expression of IL-6 mRNA in hepatocytes of all treated groups (E2, LPS and E2 & LPS hens) was elevated from 6-fold to 56-fold (P < 0.01), when compared to baseline and control values, with the highest fold change at 3 h post-treatment. The mRNA levels for IL-1β were better expressed at 24 h post-treatments with E2, LPS and E2 & LPS. The expression of IL-18 mRNA in the liver tissue was lower than IL-1β and IL-6 mRNA in all treated birds. At 24 h post-treatment, total white blood cell (WBC) counts and fibrinogen levels were elevated (P < 0.05) in E2-, LPS- and E2- & LPS-treated hens. Histologically, livers of hens from E2- and E2- & LPS-treated groups revealed inflammatory alterations with areas showing mononuclear aggregations, vacuolar fatty degeneration of hepatocytes, and increased sinusoidal congestion and haemorrhages. It was concluded that liver lipid accumulation and injury were associated with incidences of local (hepatic) and systemic inflammation, which could have initiated liver blood vessel and capsule rupture and, subsequently, the onset of FLHS.
Collapse
Affiliation(s)
- Shaniko Shini
- School of Agriculture and Food Sciences, University of Queensland, Gatton, Australia
| | - Agim Shini
- School of Agriculture and Food Sciences, University of Queensland, Gatton, Australia.,Queensland Agricultural Training Colleges (QATC), Toowoomba, Australia
| | - Wayne L Bryden
- School of Agriculture and Food Sciences, University of Queensland, Gatton, Australia
| |
Collapse
|
188
|
Kim HN, Baek JK, Park SB, Kim JD, Son HJ, Park GH, Eo HJ, Park JH, Jung HS, Jeong JB. Anti-inflammatory effect of Vaccinium oldhamii stems through inhibition of NF-κB and MAPK/ATF2 signaling activation in LPS-stimulated RAW264.7 cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:291. [PMID: 31684931 PMCID: PMC6827179 DOI: 10.1186/s12906-019-2720-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023]
Abstract
Background Vaccinium oldhamii (V. oldhamii) has been reported to exert a variety of the pharmacological properties such as anti-oxidant activity, anti-cancer activity, and inhibitory activity of α-amylase and acetylcholinesterase. However, the anti-inflammatory activity of V. oldhamii has not been studied. In this study, we aimed to investigate anti-inflammatory activity of the stem extracts from V. oldhamii, and to elucidate the potential mechanisms in LPS-stimulated RAW264.7 cells. Methods Cell viability was evaluated by MTT assay. The determination of NO and PGE2 production was performed using Griess reagent and Prostaglandin E2 ELISA Kit, respectively. The change of mRNA or protein level was evaluated by RT-PCR and Western blot. Results Among VOS, VOL and VOF, the inhibitory effect of NO and PGE2 production induced by LPS was highest in VOS treatment. Thus, VOS was selected for the further study. VOS dose-dependently blocked LPS-induced NO and PGE2 production by inhibiting iNOS and COX-2 expression, respectively. VOS inhibited the expression of pro-inflammatory cytokines such as IL-1β, IL-6 and TNF-α. In addition, VOS suppressed TRAP activity and attenuated the expression of the osteoclast-specific genes such as NFATc1, c-FOS, TRAP, MMP-9, cathepsin K, CA2, OSCAR and ATPv06d2. VOS inhibited LPS-induced NF-κB signaling activation through blocking IκB-α degradation and p65 nuclear accumulation. VOS inhibited MAPK signaling activation by attenuating the phosphorylation of ERK1/2, p38 and JNK. Furthermore, VOS inhibited ATF2 phosphorylation and blocked ATF2 nuclear accumulation. Conclusions These results indicate that VOS may exert anti-inflammatory activity by inhibiting NF-κB and MAPK/ATF2 signaling. From these findings, VOS has potential to be a candidate for the development of chemopreventive or therapeutic agents for the inflammatory diseases.
Collapse
|
189
|
Chu X, Wu S, Raju R. NLRX1 Regulation Following Acute Mitochondrial Injury. Front Immunol 2019; 10:2431. [PMID: 31736938 PMCID: PMC6830126 DOI: 10.3389/fimmu.2019.02431] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/30/2019] [Indexed: 12/17/2022] Open
Abstract
Several metabolic, cardiovascular, and neurological disorders are characterized by mitochondrial dysfunction followed by dysregulation of cellular energetics. Mitochondria play an important role in ATP production and cell death regulation. NLRX1, a mitochondria-targeted protein, is known to negatively regulate innate immunity, and cell death responses. However, the role of this protein in cellular homeostasis following mitochondrial injury is not well-understood. To understand the mechanisms underlying the effect of acute injury in regulating NLRX1 signaling pathways, we used an in vitro model of mitochondrial injury wherein, rat pulmonary microvascular endothelial cells were subjected to sodium azide treatment or glucose starvation. Both sodium azide and glucose starvation activated NF-κB and TBK1 associated innate immune response. Moreover, increased TBK1, IKK, IκB, and TRAF6 were recruited to mitochondria and interacted with NLRX1. Depletion of endogenous NLRX1 resulted in exacerbated NF-κB and TBK1 associated innate immune response and apoptosis. Our results suggest that NLRX1 participates in the regulation of innate immune response in mitochondria, and plays an important role in the maintenance of cellular homeostasis following acute mitochondrial injury. We propose that the mitochondrial recruitment of inflammatory mediators and their interaction with NLRX1 are protective responses to maintain cellular homeostasis following injury.
Collapse
Affiliation(s)
- Xiaogang Chu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Songwei Wu
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Raghavan Raju
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
190
|
Dysregulated liver lipid metabolism and innate immunity associated with hepatic steatosis in neonatal BBdp rats and NOD mice. Sci Rep 2019; 9:14594. [PMID: 31601915 PMCID: PMC6787248 DOI: 10.1038/s41598-019-51143-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
In a previous study we reported that prediabetic rats have a unique gene signature that was apparent even in neonates. Several of the changes we observed, including enhanced expression of pro-inflammatory genes and dysregulated UPR and metabolism genes were first observed in the liver followed by the pancreas. In the present study we investigated further early changes in hepatic innate immunity and metabolism in two models of type 1 diabetes (T1D), the BBdp rat and NOD mouse. There was a striking increase in lipid deposits in liver, particularly in neonatal BBdp rats, with a less striking but significant increase in neonatal NOD mice in association with dysregulated expression of lipid metabolism genes. This was associated with a decreased number of extramedullary hematopoietic clusters as well as CD68+ macrophages in the liver of both models. In addition, PPARɣ and phosphorylated AMPKα protein were decreased in neonatal BBdp rats. BBdp rats displayed decreased expression of antimicrobial genes in neonates and decreased M2 genes at 30 days. This suggests hepatic steatosis could be a common early feature in development of T1D that impacts metabolic homeostasis and tolerogenic phenotype in the prediabetic liver.
Collapse
|
191
|
Guo Q, Zheng H, Liu X, Chi S, Xu Z, Wang Q. Nutrient sensing signaling functions as the sensor and regulator of immunometabolic changes in grass carp during Flavobacteriumcolumnare infection. FISH & SHELLFISH IMMUNOLOGY 2019; 93:278-287. [PMID: 31349013 DOI: 10.1016/j.fsi.2019.07.041] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/25/2019] [Accepted: 07/14/2019] [Indexed: 06/10/2023]
Abstract
In order to illustrate the immunometabolic changes of fish during bacterial infection, grass carp (Ctenopharyngodon idellus) was injected with Flavobacteriumcolumnare(F.columnare) and then the immune response, nutrient metabolism and related signaling pathways were assayed from 6 h post injection (hpi) to 7 days post injection (dpi). After F.columnare injection, gill lamellae showed obvious fusion and higher mRNA expression levels of pro-inflammatory cytokines. The mRNA expression levels of TNF-α, IL-1β and IL-8 in the head kidney were also significantly upregulated at 6 hpi and 3 dpi. Moreover, the expression of IgZ in the gill was significantly upregulated at 3 dpi and 7 dpi, while the expression of IgM in the head kidney was significantly upregulated at 1 dpi and 3 dpi after F.columnare injection. During bacterial infection, the systematic nutrient metabolism was also significantly affected. Hepatic glycolysis, indicated by GK mRNA expression and PK activity, was significantly upregulated at 1 dpi, while glucogenesis, indicated by PEPCK mRNA expression and enzyme activity, was significantly increased at later time, which resulted in the decreased hepatic glycogen content at 1dpi but increased glycogen content at 7 dpi in the experimental group. LPL, which catalyzed the lipid catabolism, showed decreased mRNA expression and enzyme activity at 6 hpi, while ACC, which was rate-limiting of FA synthesis, was significantly increased at 6 hpi, 3 dpi and 7 dpi. During this process, the nutrient sensing signaling was also significantly affected. TOR signaling in grass carp was significantly activated while ERK signaling was significantly inhibited after F.columnare infection, both of which might function as the sensor and regulator of fish immunometabolic changes.
Collapse
Affiliation(s)
- Qian Guo
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Haiou Zheng
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Xun Liu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Shuyan Chi
- Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Zhen Xu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Qingchao Wang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| |
Collapse
|
192
|
Golia MT, Poggini S, Alboni S, Garofalo S, Ciano Albanese N, Viglione A, Ajmone-Cat MA, St-Pierre A, Brunello N, Limatola C, Branchi I, Maggi L. Interplay between inflammation and neural plasticity: Both immune activation and suppression impair LTP and BDNF expression. Brain Behav Immun 2019; 81:484-494. [PMID: 31279682 DOI: 10.1016/j.bbi.2019.07.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 06/29/2019] [Accepted: 07/03/2019] [Indexed: 01/11/2023] Open
Abstract
An increasing number of studies show that both inflammation and neural plasticity act as key players in the vulnerability and recovery from psychiatric disorders and neurodegenerative diseases. However, the interplay between these two players has been limitedly explored. In fact, while a few studies reported an immune activation, others conveyed an immune suppression, associated with an impairment in neural plasticity. Therefore, we hypothesized that deviations in inflammatory levels in both directions may impair neural plasticity. We tested this hypothesis experimentally, by acute treatment of C57BL/6 adult male mice with different doses of two inflammatory modulators: lipopolysaccharide (LPS), an endotoxin, and ibuprofen (IBU), a nonselective cyclooxygenase inhibitor, which are respectively a pro- and an anti-inflammatory agent. The results showed that LPS and IBU have different effects on behavior and inflammatory response. LPS treatment induced a reduction of body temperature, a decrease of body weight and a reduced food and liquid intake. In addition, it led to increased levels of inflammatory markers expression, both in the total hippocampus and in isolated microglia cells, including Interleukin (IL)-1β, and enhanced the concentration of prostaglandin E2 (PGE2). On the other hand, IBU increased the level of anti-inflammatory markers, decreased tryptophan 2,3-dioxygenase (TDO2), the first step in the kynurenine pathway known to be activated during inflammatory conditions, and PGE2 levels. Though LPS and IBU administration differently affected mediators related with pro- or anti-inflammatory responses, they produced overlapping effects on neural plasticity. Indeed, higher doses of both LPS and IBU induced a statistically significant decrease in the amplitude of long-term potentiation (LTP), in Brain-Derived Neurotrophic Factor (BDNF) expression levels and in the phosphorylation of the AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptor subunit GluR1, compared to the control group. Such effect appears to be dose-dependent since only the higher, but not the lower, dose of both compounds led to a plasticity impairment. Overall, the present findings indicate that acute treatment with pro- and anti-inflammatory agents impair neural plasticity in a dose dependent manner.
Collapse
Affiliation(s)
- Maria Teresa Golia
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur-Italy, Sapienza University of Rome, Italy
| | - Silvia Poggini
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Silvia Alboni
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefano Garofalo
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur-Italy, Sapienza University of Rome, Italy
| | - Naomi Ciano Albanese
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Aurelia Viglione
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy; PhD Program in Neuroscience, Scuola Superiore di Pisa, Pisa, Italy
| | | | - Abygaël St-Pierre
- Axe Neurosciences, Centre de recherche du CHU de Québec, Québec, Canada
| | - Nicoletta Brunello
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur-Italy, Sapienza University of Rome, Italy; IRCCS Neuromed, Pozzilli, Isernia, Italy
| | - Igor Branchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur-Italy, Sapienza University of Rome, Italy.
| |
Collapse
|
193
|
Abstract
Metabolic control systems coordinate myriad processes across the cellular, tissue and organismal levels to optimize the allocation of limited supplies across multiple, often competing, metabolic demands. As such, the regulation of metabolism can be analysed from the perspective of the economic theory of supply and demand. Here, we discuss how such analyses can provide new insights into the logic of metabolic control. In particular, we suggest that, in addition to being subject to well-appreciated homeostatic control, metabolism is subject to supply-driven and demand-driven controls, each operated by a dedicated set of signals throughout various physiological states, including inflammation. Furthermore, we argue that systemic homeostasis is a derived feature that evolved from the control systems that monitor metabolic supply and demand.
Collapse
Affiliation(s)
- Jessica Ye
- Howard Hughes Medical Institute and Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ruslan Medzhitov
- Howard Hughes Medical Institute and Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
194
|
Abstract
Nutrient overload occurs worldwide as a consequence of the modern diet pattern and the physical inactivity that sometimes accompanies it. Cells initiate multiple protective mechanisms to adapt to elevated intracellular metabolites and restore metabolic homeostasis, but irreversible injury to the cells can occur in the event of prolonged nutrient overload. Many studies have advanced the understanding of the different detrimental effects of nutrient overload; however, few reports have made connections and given the full picture of the impact of nutrient overload on cellular metabolism. In this review, detailed changes in metabolic and energy homeostasis caused by chronic nutrient overload, as well as their associations with the development of metabolic disorders, are discussed. Overnutrition-induced changes in key organelles and sensors rewire cellular bioenergetic pathways and facilitate the shift of the metabolic state toward biosynthesis, thereby leading to the onset of various metabolic disorders, which are essentially the downstream manifestations of a misbalanced metabolic equilibrium. Based on these mechanisms, potential therapeutic targets for metabolic disorders and new research directions are proposed.
Collapse
Affiliation(s)
- Haowen Qiu
- Department of Nutrition and Health Sciences and Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Vicki Schlegel
- Department of Nutrition and Health Sciences and Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
195
|
Amen OM, Sarker SD, Ghildyal R, Arya A. Endoplasmic Reticulum Stress Activates Unfolded Protein Response Signaling and Mediates Inflammation, Obesity, and Cardiac Dysfunction: Therapeutic and Molecular Approach. Front Pharmacol 2019; 10:977. [PMID: 31551782 PMCID: PMC6747043 DOI: 10.3389/fphar.2019.00977] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/31/2019] [Indexed: 12/25/2022] Open
Abstract
Obesity has been implicated as a risk factor for insulin resistance and cardiovascular diseases (CVDs). Although the association between obesity and CVD is a well-established phenomenon, the precise mechanisms remain incompletely understood. This has led to a relative paucity of therapeutic measures for the prevention and treatment of CVD and associated metabolic disorders. Recent studies have shed light on the pivotal role of prolonged endoplasmic reticulum stress (ERS)-initiated activation of the unfolded protein response (UPR), the ensuing chronic low-grade inflammation, and altered insulin signaling in promoting obesity-compromised cardiovascular system (CVS). In this aspect, potential ways of attenuating ERS-initiated UPR signaling seem a promising avenue for therapeutic interventions. We review intersecting role of obesity-induced ERS, chronic inflammation, insulin resistance, and oxidative stress in the discovery of targeted therapy. Moreover, this review highlights the current progress and strategies on therapeutics being explored in preclinical and clinical research to modulate ERS and UPR signaling.
Collapse
Affiliation(s)
- Omar Mohammed Amen
- School of Bioscience, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Malaysia
| | - Satyajit D. Sarker
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Reena Ghildyal
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Canberra, Australia
| | - Aditya Arya
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
- Department of Pharmacology and Therapeutics, School of Medicine, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Malaysia
- Malaysian Institute of Pharmaceuticals and Nutraceuticals, Bukit Gambir, Malaysia
| |
Collapse
|
196
|
Donath MY, Dinarello CA, Mandrup-Poulsen T. Targeting innate immune mediators in type 1 and type 2 diabetes. Nat Rev Immunol 2019; 19:734-746. [PMID: 31501536 DOI: 10.1038/s41577-019-0213-9] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2019] [Indexed: 02/07/2023]
Abstract
Type 1 and type 2 diabetes are characterized by chronic inflammation; both diseases involve pancreatic islet inflammation, while systemic low-grade inflammation is a feature of obesity and type 2 diabetes. Long-term activation of the innate immune system impairs insulin secretion and action, and inflammation also contributes to macrovascular and microvascular complications of diabetes. However, despite strong preclinical evidence and proof-of-principle clinical trials demonstrating that targeting inflammatory pathways can prevent cardiovascular disease and other complications in patients with diabetes, there are still no approved treatments for diabetes that target innate immune mediators. Here, we review recent advances in our understanding of the inflammatory pathogenesis of type 1 and type 2 diabetes from a translational angle and point out the critical gaps in knowledge that need to be addressed to guide drug development.
Collapse
Affiliation(s)
- Marc Y Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland. .,Department of Biomedicine, University of Basel, Basel, Switzerland.
| | | | | |
Collapse
|
197
|
Pattern Recognition Receptor-Mediated Chronic Inflammation in the Development and Progression of Obesity-Related Metabolic Diseases. Mediators Inflamm 2019; 2019:5271295. [PMID: 31582899 PMCID: PMC6754942 DOI: 10.1155/2019/5271295] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
Obesity-induced chronic inflammation is known to promote the development of many metabolic diseases, especially insulin resistance, type 2 diabetes mellitus, nonalcoholic fatty liver disease, and atherosclerosis. Pattern recognition receptor-mediated inflammation is an important determinant for the initiation and progression of these metabolic diseases. Here, we review the major features of the current understanding with respect to obesity-related chronic inflammation in metabolic tissues, focus on Toll-like receptors and nucleotide-binding oligomerization domain-like receptors with an emphasis on how these receptors determine metabolic disease progression, and provide a summary on the development and progress of PRR antagonists for therapeutic intervention.
Collapse
|
198
|
Coppola T, Beraud-Dufour S, Lebrun P, Blondeau N. Bridging the Gap Between Diabetes and Stroke in Search of High Clinical Relevance Therapeutic Targets. Neuromolecular Med 2019; 21:432-444. [PMID: 31489567 DOI: 10.1007/s12017-019-08563-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/13/2019] [Indexed: 12/20/2022]
Abstract
Diabetes affects more than 425 million people worldwide, a scale approaching pandemic proportion. Diabetes represents a major risk factor for stroke, and therefore is actively addressed for stroke prevention. However, how diabetes affects stroke severity has not yet been extensively considered, which is surprising given the evident but understudied common mechanistic features of both pathologies. The increase in number of diabetic people, incidence of stroke in the presence of this specific risk factor, and the exacerbation of ischemic brain damage in diabetic conditions (at least in animal models) warrants the need to integrate this comorbidity in preclinical studies of brain ischemia to develop novel therapeutic approaches. Therefore, a better understanding of the commonalties involved in the course of both diseases would offer the promise of discovering novel neuroprotective pathways that would be more appropriated to clinical scenarios. In this article, we will review the relevant mechanisms that have been identified as common traits of both pathologies and that could be, to our knowledge, potential targets in both pathologies.
Collapse
Affiliation(s)
- Thierry Coppola
- Université Côte d'Azur, CNRS, IPMC, 660 route des Lucioles, 06560, Valbonne, France.
| | - Sophie Beraud-Dufour
- Université Côte d'Azur, CNRS, IPMC, 660 route des Lucioles, 06560, Valbonne, France
| | - Patricia Lebrun
- Université Côte d'Azur, CNRS, IPMC, 660 route des Lucioles, 06560, Valbonne, France
| | - Nicolas Blondeau
- Université Côte d'Azur, CNRS, IPMC, 660 route des Lucioles, 06560, Valbonne, France.
| |
Collapse
|
199
|
Gut microbiota and obesity-associated osteoarthritis. Osteoarthritis Cartilage 2019; 27:1257-1265. [PMID: 31146016 DOI: 10.1016/j.joca.2019.05.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 05/16/2019] [Accepted: 05/18/2019] [Indexed: 02/02/2023]
Abstract
Obesity is a well-known primary risk factor for osteoarthritis (OA). In recent decades, the biomechanics-based theoretical paradigm for the pathogenesis of obesity-associated OA has been gradually but fundamentally modified. This modification is a result of accumulating evidence that biological factors also contribute to the etiology of the disease. The gut microbiota is a complicated ecosystem that profoundly influences the health of the host and can be modulated by the combined effects of environmental stimuli and genetic factors. Recently, enteric dysbacteriosis has been identified as a causal factor in the initiation and propagation of obesity-associated OA in animal models. Gut microbes and their components, microbe-associated lipid metabolites, and OA interact at both systemic and local levels through mechanisms that involve interplay with the innate immune system. However, the demonstration of causality in humans will require further studies. Nonetheless, probiotics, prebiotics, dietary habits and exercise, which aid the restoration of a healthy microbial community, are potential therapeutic approaches in the treatment of obesity-associated OA.
Collapse
|
200
|
Cao C, Wu R, Zhu X, Li Y, Li M, An F, Wu J. Ameliorative effect of Lactobacillus plantarum WW-fermented soy extract on rat fatty liver via the PPAR signaling pathway. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103439] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|