1
|
Zhang Y, Xiao B, Liu Y, Wu S, Xiang Q, Xiao Y, Zhao J, Yuan R, Xie K, Li L. Roles of PPAR activation in cancer therapeutic resistance: Implications for combination therapy and drug development. Eur J Pharmacol 2024; 964:176304. [PMID: 38142851 DOI: 10.1016/j.ejphar.2023.176304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/09/2023] [Accepted: 12/21/2023] [Indexed: 12/26/2023]
Abstract
Therapeutic resistance is a major obstacle to successful treatment or effective containment of cancer. Peroxisome proliferator-activated receptors (PPARs) play an essential role in regulating energy homeostasis and determining cell fate. Despite of the pleiotropic roles of PPARs in cancer, numerous studies have suggested their intricate relationship with therapeutic resistance in cancer. In this review, we provided an overview of the roles of excessively activated PPARs in promoting resistance to modern anti-cancer treatments, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy. The mechanisms through which activated PPARs contribute to therapeutic resistance in most cases include metabolic reprogramming, anti-oxidant defense, anti-apoptosis signaling, proliferation-promoting pathways, and induction of an immunosuppressive tumor microenvironment. In addition, we discussed the mechanisms through which activated PPARs lead to multidrug resistance in cancer, including drug efflux, epithelial-to-mesenchymal transition, and acquisition and maintenance of the cancer stem cell phenotype. Preliminary studies investigating the effect of combination therapies with PPAR antagonists have suggested the potential of these antagonists in reversing resistance and facilitating sustained cancer management. These findings will provide a valuable reference for further research on and clinical translation of PPAR-targeting treatment strategies.
Collapse
Affiliation(s)
- Yanxia Zhang
- School of Medicine, The South China University of Technology, Guangzhou, 510006, China; Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Bin Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yunduo Liu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Shunhong Wu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Qin Xiang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yuhan Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Junxiu Zhao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Ruanfei Yuan
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Keping Xie
- School of Medicine, The South China University of Technology, Guangzhou, 510006, China.
| | - Linhai Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
| |
Collapse
|
2
|
Yin Y, Xie Y, Ge W, Li Y. Creeping fat formation and interaction with intestinal disease in Crohn's disease. United European Gastroenterol J 2022; 10:1077-1084. [PMID: 36507842 PMCID: PMC9752293 DOI: 10.1002/ueg2.12349] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
Creeping fat (CrF), also known as fat wrapping, is a significant disease characteristic of Crohn's disease (CD). The transmural inflammation impairs intestinal integrity and facilitates bacteria translocation, aggravating immune response. CrF is a rich source of pro-inflammatory and pro-fibrotic cytokines with complex immune microenvironment. The inflamed and stricturing intestine is often wrapped by CrF, and CrF is associated with greater severity of CD. The large amount of innate and adaptive immune cells as well as adipocytes in CrF promote fibrosis in the affected intestine by secreting large amount of pro-fibrotic cytokines, adipokines, growth factors and fatty acids. CrF is a potential therapeutic target for CD treatment and a promising bio-marker for predicting response to drug therapy. This review aims to summarize and update the clinical manifestation and application of CrF and the underlying molecular mechanism involved in the pathogenesis of intestinal inflammation and fibrosis in CD.
Collapse
Affiliation(s)
- Yi Yin
- Department of General SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Ying Xie
- Department of General SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Wei Ge
- Department of General SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Yi Li
- Department of General SurgeryJinling HospitalMedical School of Nanjing UniversityNanjingChina
| |
Collapse
|
3
|
The Role of Transcription Factor PPAR-γ in the Pathogenesis of Psoriasis, Skin Cells, and Immune Cells. Int J Mol Sci 2022; 23:ijms23179708. [PMID: 36077103 PMCID: PMC9456565 DOI: 10.3390/ijms23179708] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/22/2022] Open
Abstract
The peroxisome proliferator-activated receptor PPAR-γ is one of three PPAR nuclear receptors that act as ligand-activated transcription factors. In immune cells, the skin, and other organs, PPAR-γ regulates lipid, glucose, and amino acid metabolism. The receptor translates nutritional, pharmacological, and metabolic stimuli into the changes in gene expression. The activation of PPAR-γ promotes cell differentiation, reduces the proliferation rate, and modulates the immune response. In the skin, PPARs also contribute to the functioning of the skin barrier. Since we know that the route from identification to the registration of drugs is long and expensive, PPAR-γ agonists already approved for other diseases may also represent a high interest for psoriasis. In this review, we discuss the role of PPAR-γ in the activation, differentiation, and proliferation of skin and immune cells affected by psoriasis and in contributing to the pathogenesis of the disease. We also evaluate whether the agonists of PPAR-γ may become one of the therapeutic options to suppress the inflammatory response in lesional psoriatic skin and decrease the influence of comorbidities associated with psoriasis.
Collapse
|
4
|
Suau R, Pardina E, Domènech E, Lorén V, Manyé J. The Complex Relationship Between Microbiota, Immune Response and Creeping Fat in Crohn's Disease. J Crohns Colitis 2022; 16:472-489. [PMID: 34528668 DOI: 10.1093/ecco-jcc/jjab159] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the last decade, there has been growing interest in the pathological involvement of hypertrophic mesenteric fat attached to the serosa of the inflamed intestinal segments involved in Crohn's disease [CD], known as creeping fat. In spite of its protective nature, creeping fat harbours an aberrant inflammatory activity which, in an already inflamed intestine, may explain why creeping fat is associated with a greater severity of CD. The transmural inflammation of CD facilitates the interaction of mesenteric fat with translocated intestinal microorganisms, contributing to activation of the immune response. This may be not the only way in which microorganisms alter the homeostasis of this fatty tissue: intestinal dysbiosis may also impair xenobiotic metabolism. All these CD-related alterations have a functional impact on nuclear receptors such as the farnesoid X receptor or the peroxisome proliferator-activated receptor γ, which are implicated in regulation of the immune response, adipogenesis and the maintenance of barrier function, as well as on creeping fat production of inflammatory-associated cells such as adipokines. The dysfunction of creeping fat worsens the inflammatory course of CD and may favour intestinal fibrosis and fistulizing complications. However, our current knowledge of the pathophysiology and pathogenic role of creeping fat is controversial and a better understanding might provide new therapeutic targets for CD. Here we aim to review and update the key cellular and molecular alterations involved in this inflammatory process that link the pathological components of CD with the development of creeping fat.
Collapse
Affiliation(s)
- Roger Suau
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| | - Eva Pardina
- Biochemistry and Molecular Biomedicine Department, University of Barcelona, Barcelona (Catalonia), Spain
| | - Eugeni Domènech
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain.,Gastroenterology Department, 'Germans Trias i Pujol' University Hospital, Badalona (Catalonia), Spain
| | - Violeta Lorén
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| | - Josep Manyé
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| |
Collapse
|
5
|
Kurzyńska A, Kunicka Z, Mierzejewski K, Golubska M, Bogacka I. PPARγ ligands modulate the immune response mediators in the pig myometrium - An in vitro study. Anim Reprod Sci 2021; 234:106866. [PMID: 34626868 DOI: 10.1016/j.anireprosci.2021.106866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 10/20/2022]
Abstract
The current study was conducted with the aim to investigate effects of PPARγ ligands on synthesis of nuclear receptor κB (NF-κB) and selected cytokines (IL-1β, IFNγ, TNFα, IL-4, IL-10, LIF) in the pig myometrium on days 14-15 of the estrous cycle (late-luteal phase) and days 14-15 of the gestational period (beginning of embryonic implantation). The myometrial slices were incubated in vitro for 6 h in medium containing PPARγ ligands, agonists: 15d-prostaglandin J2 or pioglitazone, and antagonist - T0070907. The mRNA transcript and protein abundances were evaluated in tissues and culture medium. During the estrous cycle, PPARγ ligands did not have an effect on the mRNA transcript abundance of the immune response mediators used for treatments. The IL-10 protein abundance in the tissue was less when there was inclusions of pioglitazone in the medium, while the treatment with T0070907 resulted in a larger abundance of NF-κB, IL-1β (in the tissue) and IL-4 (in tissue and culture media). During the gestational period, pioglitazone or PGJ2 suppressed mRNA IFNγ and IL-10 transcript and protein abundances (in the tissue and culture media), whereas there was an enhanced NF-κB protein abundance (in the tissue). Treatment with T0070907 had diverse effects (e.g., for NFκB inhibited mRNA transcript abundance or enhanced protein abundance). The observed changes are related mainly in tissues from pregnant animals. Responses to PPARγ antagonist are indicative of the possible involvement of PPARγ-independent factors as well as ligand-independent activation of the receptor, ligand selectivity/functionality or tissue receptivity to the factors evaluated.
Collapse
Affiliation(s)
- Aleksandra Kurzyńska
- Department of Animal Anatomy and Physiology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland.
| | - Zuzanna Kunicka
- Department of Animal Anatomy and Physiology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland
| | - Karol Mierzejewski
- Department of Animal Anatomy and Physiology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland
| | - Monika Golubska
- Department of Animal Anatomy and Physiology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland
| | - Iwona Bogacka
- Department of Animal Anatomy and Physiology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland
| |
Collapse
|
6
|
Barna BP, Malur A, Thomassen MJ. Studies in a Murine Granuloma Model of Instilled Carbon Nanotubes: Relevance to Sarcoidosis. Int J Mol Sci 2021; 22:ijms22073705. [PMID: 33918196 PMCID: PMC8038141 DOI: 10.3390/ijms22073705] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/25/2021] [Accepted: 03/31/2021] [Indexed: 12/23/2022] Open
Abstract
Poorly soluble environmental antigens, including carbon pollutants, are thought to play a role in the incidence of human sarcoidosis, a chronic inflammatory granulomatous disease of unknown causation. Currently, engineered carbon products such as multiwall carbon nanotubes (MWCNT) are manufactured commercially and have been shown to elicit acute and chronic inflammatory responses in experimental animals, including the production of granulomas or fibrosis. Several years ago, we hypothesized that constructing an experimental model of chronic granulomatosis resembling that associated with sarcoidosis might be achieved by oropharyngeal instillation of MWCNT into mice. This review summarizes the results of our efforts to define mechanisms of granuloma formation and identify potential therapeutic targets for sarcoidosis. Evidence is presented linking findings from the murine MWCNT granuloma model to sarcoidosis pathophysiology. As our goal was to determine what pulmonary inflammatory pathways might be involved, we utilized mice of knock-out (KO) backgrounds which corresponded to deficiencies noted in sarcoidosis patients. A primary example of this approach was to study mice with a myeloid-specific knock-out of the lipid-regulated transcription factor, peroxisome proliferator-activated receptor gamma (PPARγ) which is strikingly depressed in sarcoidosis. Among the major findings associated with PPARγ KO mice compared to wild-type were: (1) exacerbation of granulomatous and fibrotic histopathology in response to MWCNT; (2) elevation of inflammatory mediators; and (3) pulmonary retention of a potentially antigenic ESAT-6 peptide co-instilled with MWCNT. In line with these data, we also observed that activation of PPARγ in wild-type mice by the PPARγ-specific ligand, rosiglitazone, significantly reduced both pulmonary granuloma and inflammatory mediator production. Similarly, recognition of a deficiency of ATP-binding cassette (ABC) lipid transporter ABCG1 in sarcoidosis led us to study MWCNT instillation in myeloid-specific ABCG1 KO mice. As anticipated, ABCG1 deficiency was associated with larger granulomas and increased levels of inflammatory mediators. Finally, a transcriptional survey of alveolar macrophages from MWCNT-instilled wild-type mice and human sarcoidosis patients revealed several common themes. One of the most prominent mediators identified in both human and mouse transcriptomic analyses was MMP12. Studies with MMP12 KO mice revealed similar acute reactions to those in wild-type but at chronic time points where wild-type maintained granulomatous disease, resolution occurred with MMP12 KO mice suggesting MMP12 is necessary for granuloma progression. In conclusion, these studies suggest that the MWCNT granuloma model has relevance to human sarcoidosis study, particularly with respect to immune-specific pathways.
Collapse
|
7
|
Cao J, Ding K, Pan G, Rosario R, Su Y, Bao Y, Zhou H, Xu J, McGee Lawrence ME, Hamrick MW, Isales CM, Shi X. Deletion of PPARγ in Mesenchymal Lineage Cells Protects Against Aging-Induced Cortical Bone Loss in Mice. J Gerontol A Biol Sci Med Sci 2021; 75:826-834. [PMID: 32060555 DOI: 10.1093/gerona/glaa049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Indexed: 02/06/2023] Open
Abstract
Bone loss in aging is linked with chronic low-grade inflammation and the accumulation of marrowfat in animals and humans. Peroxisome proliferator-activated receptor gamma (PPARγ), an adipogenic regulator, plays key roles in these biological processes. However, studies of the roles of PPARγ in age-related bone loss and inflammation are lacking. We hypothesized that deletion of PPARγ in bone marrow mesenchymal lineage cells would reduce bone loss with aging, potentially through a reduction in fat-generated inflammatory responses and an increase in osteoblastic activity. In the present study, we show that mice deficient of PPARγ in Dermo1-expressing mesenchymal lineage cells (Dermo1-Cre:PPARγ fl/fl) have reduced fat mass and increased cortical bone thickness but that deficiency of PPARγ had limited effect on protection of trabecular bone with aging as demonstrated by dual-energy X-ray absorptiometry, µCT, and histomorphometric analyses. Conditional knockout of PPARγ reduced serum concentrations of adipokines, including adiponectin, resistin, and leptin, and reduced marrow stromal cell expression levels of inflammation-related genes. Inflammation genes involved in the interferon signaling pathway were reduced the most. These results demonstrate that disruption of the master adipogenic regulator, PPARγ, has a certain protective effect on aging-induced bone loss, suggesting that regulation of adipose function and modulation of interferon signaling are among the key mechanisms by which PPARγ regulates bone homeostasis during aging process.
Collapse
Affiliation(s)
- Jay Cao
- USDA-ARS Grand Forks Human Nutrition Research Center, Grand Forks, North Dakota
| | - Kehong Ding
- Center for Healthy Aging, Augusta University, Georgia.,Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Guodong Pan
- Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Raysa Rosario
- Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Yun Su
- Center for Healthy Aging, Augusta University, Georgia.,Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Yonggang Bao
- Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Hongyan Zhou
- Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Jianru Xu
- Center for Healthy Aging, Augusta University, Georgia.,Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia
| | - Meghan E McGee Lawrence
- Center for Healthy Aging, Augusta University, Georgia.,Department of Cell Biology and Anatomy, Augusta University, Georgia
| | - Mark W Hamrick
- Center for Healthy Aging, Augusta University, Georgia.,Department of Cell Biology and Anatomy, Augusta University, Georgia
| | - Carlos M Isales
- Center for Healthy Aging, Augusta University, Georgia.,Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia.,Department of Medicine, Augusta University, Georgia
| | - Xingming Shi
- Center for Healthy Aging, Augusta University, Georgia.,Department of Neuroscience and Regenerative Medicine, Augusta University, Georgia.,Department of Medicine, Augusta University, Georgia
| |
Collapse
|
8
|
Hernandez-Quiles M, Broekema MF, Kalkhoven E. PPARgamma in Metabolism, Immunity, and Cancer: Unified and Diverse Mechanisms of Action. Front Endocrinol (Lausanne) 2021; 12:624112. [PMID: 33716977 PMCID: PMC7953066 DOI: 10.3389/fendo.2021.624112] [Citation(s) in RCA: 194] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/08/2021] [Indexed: 12/20/2022] Open
Abstract
The proliferator-activated receptor γ (PPARγ), a member of the nuclear receptor superfamily, is one of the most extensively studied ligand-inducible transcription factors. Since its identification in the early 1990s, PPARγ is best known for its critical role in adipocyte differentiation, maintenance, and function. Emerging evidence indicates that PPARγ is also important for the maturation and function of various immune system-related cell types, such as monocytes/macrophages, dendritic cells, and lymphocytes. Furthermore, PPARγ controls cell proliferation in various other tissues and organs, including colon, breast, prostate, and bladder, and dysregulation of PPARγ signaling is linked to tumor development in these organs. Recent studies have shed new light on PPARγ (dys)function in these three biological settings, showing unified and diverse mechanisms of action. Classical transactivation-where PPARγ activates genes upon binding to PPAR response elements as a heterodimer with RXRα-is important in all three settings, as underscored by natural loss-of-function mutations in FPLD3 and loss- and gain-of-function mutations in tumors. Transrepression-where PPARγ alters gene expression independent of DNA binding-is particularly relevant in immune cells. Interestingly, gene translocations resulting in fusion of PPARγ with other gene products, which are unique to specific carcinomas, present a third mode of action, as they potentially alter PPARγ's target gene profile. Improved understanding of the molecular mechanism underlying PPARγ activity in the complex regulatory networks in metabolism, cancer, and inflammation may help to define novel potential therapeutic strategies for prevention and treatment of obesity, diabetes, or cancer.
Collapse
Affiliation(s)
- Miguel Hernandez-Quiles
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Marjoleine F. Broekema
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Eric Kalkhoven
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- *Correspondence: Eric Kalkhoven,
| |
Collapse
|
9
|
Bae HR, Choi MS, Kim S, Young HA, Gershwin ME, Jeon SM, Kwon EY. IFNγ is a Key Link between Obesity and Th1-Mediated AutoImmune Diseases. Int J Mol Sci 2020; 22:ijms22010208. [PMID: 33379198 PMCID: PMC7794719 DOI: 10.3390/ijms22010208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/22/2022] Open
Abstract
Obesity, a characteristic of metabolic syndrome, is also associated with chronic inflammation and the development of autoimmune diseases. However, the relationship between obesity and autoimmune diseases remains to be investigated in depth. Here, we compared hepatic gene expression profiles among high-fat diet (HFD) mice using the primary biliary cholangitis (PBC) mouse model based on the chronic expression of interferon gamma (IFNγ) (ARE-Del-/- mice). The top differentially expressed genes affected by upstream transcriptional regulators IFNγ, LPS, and TNFα displayed an overlap in HFD and ARE-Del-/- mice, indicating that obesity-induced liver inflammation may be dependent on signaling via IFNγ. The top pathways altered in HFD mice were mostly involved in the innate immune responses, which overlapped with ARE-Del-/- mice. In contrast, T cell-mediated signaling pathways were exclusively altered in ARE-Del-/- mice. We further evaluated the therapeutic effect of luteolin, known as anti-inflammatory flavonoid, in HFD and ARE-Del-/- mice. Luteolin strongly suppressed the MHC I and II antigen presentation pathways, which were highly activated in both HFD and ARE-Del-/- mice. Conversely, luteolin increased metabolic processes of fatty acid oxidation and oxidative phosphorylation in the liver, which were suppressed in ARE-Del-/- mice. Luteolin also strongly induced PPAR signaling, which was downregulated in HFD and ARE-Del-/- mice. Using human GWAS data, we characterized the genetic interaction between significant obesity-related genes and IFNγ signaling and demonstrated that IFNγ is crucial for obesity-mediated inflammatory responses. Collectively, this study improves our mechanistic understanding of the relationship between obesity and autoimmune diseases. Furthermore, it provides new methodological insights into how immune network-based analyses effectively integrate RNA-seq and microarray data.
Collapse
Affiliation(s)
- Heekyong R. Bae
- Omixplus, LLC., Gaithersburg, MD 20885, USA; (H.R.B.); (S.K.)
- Laboratory of Cancer Immunometabolism, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21701, USA;
- Department of Food Science and Nutrition, Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu 41566, Korea;
| | - Myung-Sook Choi
- Department of Food Science and Nutrition, Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu 41566, Korea;
| | - Suntae Kim
- Omixplus, LLC., Gaithersburg, MD 20885, USA; (H.R.B.); (S.K.)
| | - Howard A. Young
- Laboratory of Cancer Immunometabolism, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21701, USA;
| | - M. Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616, USA;
| | - Seon-Min Jeon
- R&D Center, APtechnologies Corp., Gyeonggi-do, Hwaseong-si 18469, Korea
- Correspondence: (S.-M.J.); (E.-Y.K.); Tel.: +82-53-950-7936 (S.-M.J.); +82-53-950-6231 (E.-Y.K.)
| | - Eun-Young Kwon
- Department of Food Science and Nutrition, Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu 41566, Korea;
- Correspondence: (S.-M.J.); (E.-Y.K.); Tel.: +82-53-950-7936 (S.-M.J.); +82-53-950-6231 (E.-Y.K.)
| |
Collapse
|
10
|
Sarkar P, Thirumurugan K. New insights into TNFα/PTP1B and PPARγ pathway through RNF213- a link between inflammation, obesity, insulin resistance, and Moyamoya disease. Gene 2020; 771:145340. [PMID: 33333224 DOI: 10.1016/j.gene.2020.145340] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 11/11/2020] [Accepted: 12/01/2020] [Indexed: 01/02/2023]
Abstract
Diabetic patients are always at a higher risk of ischemic diseases like coronary artery diseases. One such ischemic carotid artery disease is Moyamoya disease (MMD) associated with diabetes Type I and II, but the causality was unclear. Ring Finger Protein 213 (RNF213) is the major susceptible gene for MMD. To understand the association between diabetes mellitus and MMD we chose the major players from both of the anomalies: insulin and RNF213. But before establishing the role of RNF213 in the insulin-regulating pathway we had to understand the involvement of RNF213 within different biological systems. For this, we have adopted a preliminary computational approach to find the prominent interactions of RNF213. Our first objective was to construct an interactome for RNF213. We have analyzed several curated databases and adapted a list of RNF213 interacting partners to develop its interactome. Then to understand the involvement of this interactome in biological functions we have analyzed major biological pathways, biological processes, and prominent clusters related to this interactome through a computational approach. Then to develop a pathway that might give clues for RNF213 involvement in the insulin regulatory pathway we have validated the intercluster and intracluster predictions and identified a regulatory pathway for RNF213. RNF213 interactome was observed to be involved in adaptive immunity with 4 major clusters; one of the clusters involved TNFα. The immune system involves several pathways, and therefore at this point, we have chosen an event-based strategy to obtain an explicit target. Immunity is mediated by pro-inflammatory cytokines like TNFα. TNFα-mediated inflammation, obesity, and insulin resistance are associated. Therefore we chose to explore the role of RNF213 in TNFα-mediated inflammation in macrophages and inflammation-mediated insulin-resistance in adipocytes. We have observed an enhancement of RNF213 gene expression by LPS mediated pro-inflammatory stimuli and suppression by PPARγ-mediated anti-inflammatory, insulin-sensitizing stimuli in macrophages, and also in adipocytes. Administration of the pro-inflammatory cytokine TNFα was able to impede the reduction in RNF213 expression during adipogenesis and this effect was observed to be mediated by PTP1B. Inactivation of PTP1B abolished RNF213 expression which in turn enhanced the adipogenesis process through enhanced PPARγ. Constitutive expression of RNF213 suppressed the adipocyte differentiation by the inhibition of PPARγ. We could show the regulation of RNF213 by TNFα/PTP1B pathway and PPARγ. The constitutive expression of RNF213 during adipogenesis appears to be an adipostatic measure that obese patients acquire to inhibit further adipogenesis. This is verified in silico by analyzing the gene expression data obtained from the Gene Expression Omnibus database, which showed a higher expression of RNF213 in adipose tissue samples of obese people. Overall this study gives new insights into the TNFα-mediated pathway in adipogenesis and suggests the role of RNF213 in adipogenesis via this pathway.
Collapse
Affiliation(s)
- Priyanka Sarkar
- 206, Structural Biology Lab, Centre for Biomedical Research, School of Biosciences & Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Kavitha Thirumurugan
- 206, Structural Biology Lab, Centre for Biomedical Research, School of Biosciences & Technology, Vellore Institute of Technology, Vellore 632014, India.
| |
Collapse
|
11
|
Mohan A, Neequaye N, Malur A, Soliman E, McPeek M, Leffler N, Ogburn D, Tokarz DA, Knudson W, Gharib SA, Schnapp LM, Barna BP, Thomassen MJ. Matrix Metalloproteinase-12 Is Required for Granuloma Progression. Front Immunol 2020; 11:553949. [PMID: 33072094 PMCID: PMC7531023 DOI: 10.3389/fimmu.2020.553949] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022] Open
Abstract
Background Sarcoidosis is a chronic inflammatory disease of unknown cause characterized by granuloma formation. Mechanisms for chronic persistence of granulomas are unknown. Matrix Metalloproteinase-12 (MMP12) degrades extracellular matrix elastin and enables infiltration of immune cells responsible for inflammation and granuloma formation. Previous studies report increased MMP12 in sarcoidosis patients and association between MMP12 expression and disease severity. We also observed elevated MMP12 in our multiwall carbon nanotube (MWCNT) murine model of granulomatous inflammation. Here we hypothesized that MMP12 is important to acute and late phases of granuloma pathogenesis. To test this hypothesis, we analyzed granulomatous and inflammatory responses of Mmp12 knock-out (KO) mice at 10 (acute) and 60 days (late) after MWCNT instillation. Methods C57BL/6 (wildtype) and Mmp12 KO mice underwent oropharyngeal instillation of MWCNT. Lungs were harvested at 3, 10, 20, and 60 days post instillation for evaluation of MMP12 expression and granulomatous changes. Bronchoalveolar lavage (BAL) cells were analyzed 60 days after MWCNT instillation for expression of mediators thought to play a role in sarcoid granulomatosis: peroxisome proliferator-activated receptor-gamma (PPARγ), interferon-gamma (IFN-γ), and CCL2 (MCP-1). Results Pulmonary granuloma appearance at 10 days after MWCNT instillation showed no differences between wildtype and Mmp12 KO mice. In contrast, by 60 days after MWCNT instillation, Mmp12 KO mice revealed markedly attenuated granuloma formation together with elevated PPARγ and reduced IFNγ expression in BAL cells compared to wildtype. Unexpectedly, Mmp12 KO mice further demonstrated increased alveolar macrophages with increased CCL2 at 60 days. Conclusions The striking reduction of granuloma formation at day 60 in Mmp12 KO mice suggests that MMP12 is required to maintain chronic granuloma pathophysiology. The increased PPARγ and decreased IFNγ findings suggest that these mediators also may be involved since previous studies have shown that PPARγ suppresses IFNγ and PPARγ deficiency amplifies granuloma formation. Interestingly, a role of MMP12 in granuloma resolution is also suggested by increases in both macrophage influx and CCL2. Overall, our results strongly implicate MMP12 as a key factor in granuloma persistence and as a possible therapeutic target in chronic pulmonary sarcoidosis.
Collapse
Affiliation(s)
- Arjun Mohan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine- East Carolina University, Greenville, NC, United States
| | - Nicole Neequaye
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine- East Carolina University, Greenville, NC, United States
| | - Anagha Malur
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine- East Carolina University, Greenville, NC, United States
| | - Eman Soliman
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine- East Carolina University, Greenville, NC, United States.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Matthew McPeek
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine- East Carolina University, Greenville, NC, United States
| | - Nancy Leffler
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine- East Carolina University, Greenville, NC, United States
| | - David Ogburn
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine- East Carolina University, Greenville, NC, United States
| | - Debra A Tokarz
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Warren Knudson
- Department of Anatomy and Cell Biology, Brody School of Medicine- East Carolina University, Greenville, NC, United States
| | - Sina A Gharib
- Division of Pulmonary, Critical Care and Sleep Medicine, Computational Medicine Core, Center for Lung Biology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Lynn M Schnapp
- Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Barbara P Barna
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine- East Carolina University, Greenville, NC, United States
| | - Mary Jane Thomassen
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Brody School of Medicine- East Carolina University, Greenville, NC, United States
| |
Collapse
|
12
|
Fu S, He K, Tian C, Sun H, Zhu C, Bai S, Liu J, Wu Q, Xie D, Yue T, Shen Z, Dai Q, Yu X, Zhu S, Liu G, Zhou R, Duan S, Tian Z, Xu T, Wang H, Bai L. Impaired lipid biosynthesis hinders anti-tumor efficacy of intratumoral iNKT cells. Nat Commun 2020; 11:438. [PMID: 31974378 PMCID: PMC6978340 DOI: 10.1038/s41467-020-14332-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022] Open
Abstract
Dysfunction of invariant natural killer T (iNKT) cells in tumor microenvironment hinders their anti-tumor efficacy, and the underlying mechanisms remain unclear. Here we report that iNKT cells increase lipid biosynthesis after activation, and that is promoted by PPARγ and PLZF synergically through enhancing transcription of Srebf1. Among those lipids, cholesterol is required for the optimal IFN-γ production from iNKT cells. Lactic acid in tumor microenvironment reduces expression of PPARγ in intratumoral iNKT cells and consequently diminishes their cholesterol synthesis and IFN-γ production. Importantly, PPARγ agonist pioglitazone, a thiazolidinedione drug for type 2 diabetes, successfully restores IFN-γ production in tumor-infiltrating iNKT cells from both human patients and mouse models. Combination of pioglitazone and alpha-galactosylceramide treatments significantly enhances iNKT cell-mediated anti-tumor immune responses and prolongs survival of tumor-bearing mice. Our studies provide a strategy to augment the anti-tumor efficacy of iNKT cell-based immunotherapies via promoting their lipid biosynthesis. Lipid metabolism has been linked to iNKT function largely as it impacts processing and presentation of lipids they recognize. Here the authors show that iNKT-intrinsic lipid biosynthesis is important for their function but is impaired in tumors, and its restoration with PPARγ agonist drugs promotes anti-tumor iNKT response.
Collapse
Affiliation(s)
- Sicheng Fu
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Kaixin He
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Chenxi Tian
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hua Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
| | - Chenwen Zhu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
| | - Shiyu Bai
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Jiwei Liu
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Qielan Wu
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Di Xie
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ting Yue
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Zhuxia Shen
- Department of Cardiology, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, 200040, China
| | - Qingqing Dai
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xiaojun Yu
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Shu Zhu
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Gang Liu
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, 230027, China
| | - Rongbin Zhou
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Shengzhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Zhigang Tian
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Tao Xu
- National Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China.
| | - Li Bai
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China. .,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
13
|
Peroxisome Proliferator-Activated Receptor Gamma (PPAR) Suppresses Inflammation and Bacterial Clearance during Influenza-Bacterial Super-Infection. Viruses 2019; 11:v11060505. [PMID: 31159430 PMCID: PMC6630660 DOI: 10.3390/v11060505] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/28/2019] [Accepted: 05/30/2019] [Indexed: 01/21/2023] Open
Abstract
Influenza virus is among the most common causes of respiratory illness worldwide and can be complicated by secondary bacterial pneumonia, a frequent cause of mortality. When influenza virus infects the lung, the innate immune response is activated, and interferons and inflammatory mediators are released. This "cytokine storm" is thought to play a role in influenza-induced lung pathogenesis. Peroxisome proliferator-activated receptor gamma (PPARγ) is a member of the nuclear hormone receptor super-family. PPARγ has numerous functions including enhancing lipid and glucose metabolism and cellular differentiation and suppressing inflammation. Synthetic PPARγagonists (thiazolidinediones or glitazones) have been used clinically in the treatment of type II diabetes. Using data from the National Health and Nutrition Examination Survey (NHANES), diabetic participants taking rosiglitazone had an increased risk of mortality from influenza/pneumonia compared to those not taking the drug. We examined the effect of rosiglitazone treatment during influenza and secondary bacterial (Methicillin resistant Staphylococcus aureus) pneumonia in mice. We found decreased influenza viral burden, decreased numbers of neutrophils and macrophages in bronchoalveolar lavage, and decreased production of cytokines and chemokines in influenza infected, rosiglitazone-treated mice when compared to controls. However, rosiglitazone treatment compromised bacterial clearance during influenza-bacterial super-infection. Both human and mouse data suggest that rosiglitazone treatment worsens the outcome of influenza-associated pneumonia.
Collapse
|
14
|
Daryabor G, Kabelitz D, Kalantar K. An update on immune dysregulation in obesity-related insulin resistance. Scand J Immunol 2019; 89:e12747. [PMID: 30593678 DOI: 10.1111/sji.12747] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/22/2018] [Accepted: 12/25/2018] [Indexed: 12/29/2022]
Abstract
Obesity is associated with chronic low-grade inflammation of the adipose tissue (AT) that might develop into systemic inflammation, insulin resistance (IR) and an increased risk of type 2 diabetes mellitus (T2DM) in severe obese rodents and humans. In the lean state, small normal adipocytes and AT macrophages interact with each other to maintain metabolic homeostasis but during obesity, enlarged adipocytes secrete inflammatory mediators and express immune receptors to recruit immune cells and aggravate the inflammation. The better understanding of the obesity-related inflammatory milieu and the sequential events leading to IR could be helpful in designing new preventive and therapeutic strategies. The present review will discuss the cellular and molecular abnormalities participating in the pathogenesis of obesity in obese individuals as well as high-fat diet (HFD)-fed mice, a mouse model of obesity.
Collapse
Affiliation(s)
- Gholamreza Daryabor
- Department of Immunology, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Kurosh Kalantar
- Department of Immunology, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
15
|
Abstract
Interferon gamma, referred to here as IFN-γ, is a major component in immunological cell signaling and is a critical regulatory protein for overall immune system function. First discovered in 1965 (Wheelock Science 149: (3681)310-311, 1965), IFN-γ is the only Type II interferon identified. Its expression is both positively and negatively controlled by different factors. In this chapter, we will review the transcriptional and post-transcriptional control of IFN-γ expression. In the transcriptional control part, the regular activators and suppressors are summarized, we will also focus on the epigenetic control, such as chromosome access, DNA methylation, and histone acetylation. The more we learn about the control of this regulatory protein will allow us to apply this knowledge in the future to effectively manipulate IFN-γ expression for the treatment of infections, cancer, inflammation, and autoimmune diseases.
Collapse
|
16
|
Kim TH, Choi D, Kim JY, Lee JH, Koo SH. Fast food diet-induced non-alcoholic fatty liver disease exerts early protective effect against acetaminophen intoxication in mice. BMC Gastroenterol 2017; 17:124. [PMID: 29179698 PMCID: PMC5704433 DOI: 10.1186/s12876-017-0680-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 11/15/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Acetaminophen (APAP) is a readily available and safe painkiller. However, its overdose is the most common cause of acute liver injury (ALI). Many predisposing factors contribute to susceptibility to APAP-induced ALI. Non-alcoholic fatty liver disease (NAFLD), the major cause of chronic liver disease, is considered an important predictor of APAP-induced ALI, although the exact mechanism controversial. In this study, we aimed to elucidate the effects of NAFLD on APAP-induced ALI. METHODS Two groups of mice, normal chow (NC) diet-fed and fast food (FF) diet-fed mice for 14 weeks, were further divided into two subgroups: intraperitoneally injected with either saline (NC-S and FF-S groups) or APAP (NC-A and FF-A groups). Biochemical tests, histological analysis, quantitative PCR, and western blotting were conducted. RESULTS Alanine aminotransferase (ALT) level (199.0 ± 39.0 vs. 63.8 ± 7.4 IU/L, p < 0.05) and NAFLD activity score (0 vs. 4.5 ± 0.22) were significantly higher in mice in FF-S group than those in NC-S group. ALI features such as ALT level (8447.8 ± 1185.3 vs. 836.6 ± 185.1 IU/L, p < 0.001) and centrizonal necrosis were prominent and mRNA levels of Trib3 (RR, 1.81) was high in mice in the NC-A group. Levels of CYP2E1 and anti-inflammatory molecules such as PPAR-γ, p62, and NRF2 were high in mice in the FF-A group. CONCLUSIONS Our results showed that while the FF diet clearly induced non-alcoholic steatohepatitis and metabolic syndrome, NAFLD also attenuates APAP-induced ALI by inducing anti-inflammatory molecules such as PPAR-γ.
Collapse
Affiliation(s)
- Tae Hyung Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, South Korea
| | - Dahee Choi
- Division of Life Sciences, Korea University College of Life Sciences & Biotechnology, 145 Anam-Ro Seongbuk-Gu, Seoul, 02841, South Korea
| | - Joo Young Kim
- Department of Pathology, Korea University College of Medicine, Seoul, South Korea
| | - Jeong Hyeon Lee
- Department of Pathology, Korea University College of Medicine, Seoul, South Korea
| | - Seung-Hoi Koo
- Division of Life Sciences, Korea University College of Life Sciences & Biotechnology, 145 Anam-Ro Seongbuk-Gu, Seoul, 02841, South Korea.
| |
Collapse
|
17
|
Cai W, Yang T, Liu H, Han L, Zhang K, Hu X, Zhang X, Yin KJ, Gao Y, Bennett MVL, Leak RK, Chen J. Peroxisome proliferator-activated receptor γ (PPARγ): A master gatekeeper in CNS injury and repair. Prog Neurobiol 2017; 163-164:27-58. [PMID: 29032144 DOI: 10.1016/j.pneurobio.2017.10.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/06/2017] [Accepted: 10/08/2017] [Indexed: 01/06/2023]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a widely expressed ligand-modulated transcription factor that governs the expression of genes involved in inflammation, redox equilibrium, trophic factor production, insulin sensitivity, and the metabolism of lipids and glucose. Synthetic PPARγ agonists (e.g. thiazolidinediones) are used to treat Type II diabetes and have the potential to limit the risk of developing brain injuries such as stroke by mitigating the influence of comorbidities. If brain injury develops, PPARγ serves as a master gatekeeper of cytoprotective stress responses, improving the chances of cellular survival and recovery of homeostatic equilibrium. In the acute injury phase, PPARγ directly restricts tissue damage by inhibiting the NFκB pathway to mitigate inflammation and stimulating the Nrf2/ARE axis to neutralize oxidative stress. During the chronic phase of acute brain injuries, PPARγ activation in injured cells culminates in the repair of gray and white matter, preservation of the blood-brain barrier, reconstruction of the neurovascular unit, resolution of inflammation, and long-term functional recovery. Thus, PPARγ lies at the apex of cell fate decisions and exerts profound effects on the chronic progression of acute injury conditions. Here, we review the therapeutic potential of PPARγ in stroke and brain trauma and highlight the novel role of PPARγ in long-term tissue repair. We describe its structure and function and identify the genes that it targets. PPARγ regulation of inflammation, metabolism, cell fate (proliferation/differentiation/maturation/survival), and many other processes also has relevance to other neurological diseases. Therefore, PPARγ is an attractive target for therapies against a number of progressive neurological disorders.
Collapse
Affiliation(s)
- Wei Cai
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Huan Liu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lijuan Han
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kai Zhang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Xiaoming Hu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai 200032, China; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh PA, USA
| | - Xuejing Zhang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Michael V L Bennett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA.
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai 200032, China; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh PA, USA.
| |
Collapse
|
18
|
Abstract
In healthy individuals, metabolically quiescent T cells survey lymph nodes and peripheral tissues in search of cognate antigens. During infection, T cells that encounter cognate antigens are activated and - in a context-specific manner - proliferate and/or differentiate to become effector T cells. This process is accompanied by important changes in cellular metabolism (known as metabolic reprogramming). The magnitude and spectrum of metabolic reprogramming as it occurs in T cells in the context of acute infection ensure host survival. By contrast, altered T cell metabolism, and hence function, is also observed in various disease states, in which T cells actively contribute to pathology. In this Review, we introduce the idea that the spectrum of immune cell metabolic states can provide a basis for categorizing human diseases. Specifically, we first summarize the metabolic and interlinked signalling requirements of T cells responding to acute infection. We then discuss how metabolic reprogramming of T cells is linked to disease.
Collapse
|
19
|
Madapura HS, Nagy N, Ujvari D, Kallas T, Kröhnke MCL, Amu S, Björkholm M, Stenke L, Mandal PK, McMurray JS, Keszei M, Westerberg LS, Cheng H, Xue F, Klein G, Klein E, Salamon D. Interferon γ is a STAT1-dependent direct inducer of BCL6 expression in imatinib-treated chronic myeloid leukemia cells. Oncogene 2017; 36:4619-4628. [PMID: 28368400 DOI: 10.1038/onc.2017.85] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 01/20/2017] [Accepted: 02/26/2017] [Indexed: 01/12/2023]
Abstract
B-cell CLL/lymphoma 6 (BCL6) exerts oncogenic effects in several human hematopoietic malignancies including chronic myeloid leukemia (CML), where BCL6 expression was shown to be essential for CML stem cell survival and self-renewal during imatinib mesylate (IM) treatment. As several lines of evidence suggest that interferon γ (IFNγ) production in CML patients might have a central role in the response to tyrosine kinase inhibitor (TKI) therapy, we analyzed if IFNγ modulates BCL6 expression in CML cells. Although separate IFNγ or IM treatment only slightly upregulated BCL6 expression, combined treatment induced remarkable BCL6 upregulation in CML lines and primary human CD34+ CML stem cells. We proved that during combined treatment, inhibition of constitutive signal transducer and activator of transcription (STAT) 5 activation by IM allowed the specific enhancement of the STAT1 dependent, direct upregulation of BCL6 by IFNγ in CML cells. By using colony-forming assay, we found that IFNγ enhanced the ex vivo colony or cluster-forming capacity of human CML stem cells in the absence or presence of IM, respectively. Furthermore, inhibition of the transcriptional repressor function of BCL6 in the presence of IM and IFNγ almost completely blocked the cluster formation of human CML stem cells. On the other hand, by using small interfering RNA knockdown of BCL6, we demonstrated that in an IM-treated CML line the antiapoptotic effect of IFNγ was independent of BCL6 upregulation. We found that IFNγ also upregulated several antiapoptotic members of the BCL2 and BIRC gene families in CML cells, including the long isoform of MCL1, which proved to be essential for the antiapoptotic effect of IFNγ in an IM-treated CML line. Our results suggest that combination of TKIs with BCL6 and MCL1 inhibitors may potentially lead to the complete eradication of CML stem cells.
Collapse
Affiliation(s)
- H S Madapura
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - N Nagy
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - D Ujvari
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - T Kallas
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - M C L Kröhnke
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - S Amu
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - M Björkholm
- Division of Hematology, Department of Medicine, Karolinska University Hospital Solna and Karolinska Institutet, Stockholm, Sweden
| | - L Stenke
- Division of Hematology, Department of Medicine, Karolinska University Hospital Solna and Karolinska Institutet, Stockholm, Sweden
| | - P K Mandal
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - J S McMurray
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - M Keszei
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - L S Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - H Cheng
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - F Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - G Klein
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - E Klein
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - D Salamon
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Pleiotropic effects of statins: new therapeutic targets in drug design. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:695-712. [PMID: 27146293 DOI: 10.1007/s00210-016-1252-4] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
Abstract
The HMG Co-enzyme inhibitors and new lipid-modifying agents expand their new therapeutic target options in the field of medical profession. Statins have been described as the most effective class of drugs to reduce serum cholesterol levels. Since the discovery of the first statin nearly 30 years ago, these drugs have become the main therapeutic approach to lower cholesterol levels. The present scientific research demonstrates numerous non-lipid modifiable effects of statins termed as pleiotropic effects of statins, which could be beneficial for the treatment of various devastating disorders. The most important positive effects of statins are anti-inflammatory, anti-proliferative, antioxidant, immunomodulatory, neuroprotective, anti-diabetes, and antithrombotic, improving endothelial dysfunction and attenuating vascular remodeling besides many others which are discussed under the scope of this review. In particular, inhibition of Rho and its downstream target, Rho-associated coiled-coil-containing protein kinase (ROCK), and their agonistic action on peroxisome proliferator-activated receptors (PPARs) can be viewed as the principle mechanisms underlying the pleiotropic effects of statins. With gradually increasing knowledge of new therapeutic targets of statins, their use has also been advocated in chronic inflammatory disorders for example rheumatoid arthritis (RA) and in systemic lupus erythematosus (SLE). In the scope of review, we highlight statins and their pleiotropic effects with reference to their harmful and beneficial effects as a novel approach for their use in the treatment of devastating disorders. Graphical abstract Pleiotropic effect of statins.
Collapse
|
21
|
Abstract
LXR (liver X receptor) is a ligand-activated transcription factor and plays an important role in regulation of lipid homoeostasis and inflammation. Several studies indicate that LXR inhibits IFN-γ (interferon γ)-induced biological responses; however, the influence of LXR on IFN-γ expression has not been fully elucidated. In the present study, we investigated the effects of LXR activation on IFN-γ expression at different levels. At the molecular level, we surprisingly observed that LXR ligand (T0901317) induced macrophage and T-cell IFN-γ protein expression which was associated with increased mRNA and secreted protein levels in culture medium. In contrast, selective inhibition of LXRα and/or LXRβ expression by siRNA reduced IFN-γ expression. Promoter analysis defined the multiple LXREs (LXR-responsive elements) in the proximal region of the IFN-γ promoter. EMSAs and ChIP indicated that LXR activation enhanced the binding of LXR protein to these LXREs. In vivo, T0901317 increased wild-type mouse serum IFN-γ levels and IFN-γ expression in the lung and lymph nodes. Functionally, we observed that administration of T0901317 to wild-type mice increased rates of survival and being tumour-free, and inhibited tumour growth when the animals were inoculated with LLC1 carcinoma. In contrast, these protective effects were substantially attenuated in IFN-γ-knockout (IFN-γ-/-) mice, suggesting that the induction of IFN-γ production plays a critical role in T0901317-inhibited tumour growth. Taken together, the results of the present study show that IFN-γ is another molecular target of LXR activation, and it suggests a new mechanism by which LXR inhibits tumour growth.
Collapse
|
22
|
Yuan M, Qiu M, Cui J, Zhang X, Zhang P. Protective effects of pioglitazone against immunoglobulin deposition on heart of streptozotocin-induced diabetic rats. J Endocrinol Invest 2014; 37:375-84. [PMID: 24682915 DOI: 10.1007/s40618-013-0046-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 12/17/2013] [Indexed: 11/28/2022]
Abstract
AIM Peroxisome proliferator-activated receptor-γ (PPAR-γ) agonists have immunomodulatory and anti-inflammatory effects. The study investigated the autoimmune injuries of diabetic cardiomyopathy (DCM) and tested the hypothesis that PPAR-γ agonists suppress disordered immune responses in diabetic heart, thereby preventing evolution of DCM. METHODS STZ-induced diabetic rats were assigned to five groups: DM group, given no treatment; INS group, given insulin (4 U kg(-1) d(-1)); PIL group, given low dose pioglitazone (4 mg kg(-1) d(-1)); PIL/INS group, given both low dose pioglitazone and insulin; PIH group, given high dose pioglitazone (20 mg kg(-1) d(-1)). Normal rats (CON group) were also monitored as control. The pathologic abnormalities of hearts were observed. The immunoglobulin deposition was examined by immunohistochemistry and immunofluorescence. RESULTS At 16 weeks, interstitial fibrosis was shown in diabetic heart which was accompanied by plenty of inflammatory cells infiltrated. Pioglitazone therapy could ameliorate the cardiac injuries. Shown by immunohistochemistry, the difference of integrated optical density (IOD) of immunoglobulin deposition among each group had statistic significance. No obvious immunoglobulins were deposited in the intercellular substance of heart in CON group (IgA 290.8 ± 88.1, IgG 960.4 ± 316.0 and IgM 341.3 ± 67.9). But the deposition of immunoglobulins increased significantly in DM group (IgA 7,047.5 ± 1,328.3, P < 0.05; IgG 28,945.9 ± 5,160.7, P < 0.05 and IgM 8,580.8 ± 1,336.8, P < 0.05). Administration of pioglitazone greatly reduced the increased deposition in a dose-dependent fashion. Moreover, the statistical significance was the same with immunofluorescence analysis as with immunohistochemical examination. CONCLUSIONS The data suggest that disordered immune responses play an important role in the pathogenesis of DCM. Pioglitazone showed protective effects by inhibiting the immunoglobulin deposition on diabetic myocardium.
Collapse
Affiliation(s)
- M Yuan
- Department of Endocrinology, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | | | | | | | | |
Collapse
|
23
|
Schmuth M, Moosbrugger-Martinz V, Blunder S, Dubrac S. Role of PPAR, LXR, and PXR in epidermal homeostasis and inflammation. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:463-73. [PMID: 24315978 DOI: 10.1016/j.bbalip.2013.11.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 11/18/2013] [Accepted: 11/23/2013] [Indexed: 12/19/2022]
Abstract
Epidermal lipid synthesis and metabolism are regulated by nuclear hormone receptors (NHR) and in turn epidermal lipid metabolites can serve as ligands to NHR. NHR form a large superfamily of receptors modulating gene transcription through DNA binding. A subgroup of these receptors is ligand-activated and heterodimerizes with the retinoid X receptor including peroxisome proliferator-activated receptor (PPAR), liver X receptor (LXR) and pregnane X receptor (PXR). Several isotypes of these receptors exist, all of which are expressed in skin. In keratinocytes, ligand activation of PPARs and LXRs stimulates differentiation, induces lipid accumulation, and accelerates epidermal barrier regeneration. In the cutaneous immune system, ligand activation of all three receptors, PPAR, LXR, and PXR, has inhibitory properties, partially mediated by downregulation of the NF-kappaB pathway. PXR also has antifibrotic effects in the skin correlating with TGF-beta inhibition. In summary, ligands of PPAR, LXR and PXR exert beneficial therapeutic effects in skin disease and represent promising targets for future therapeutic approaches in dermatology. This article is part of a Special Issue entitled The Important Role of Lipids in the Epidermis and their Role in the Formation and Maintenance of the Cutaneous Barrier. Guest Editors: Kenneth R. Feingold and Peter Elias.
Collapse
Affiliation(s)
- Matthias Schmuth
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria.
| | | | - Stefan Blunder
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria.
| |
Collapse
|
24
|
Peroxisome proliferator-activated receptor-γ agonist pioglitazone suppresses experimental autoimmune uveitis. Exp Eye Res 2013; 116:291-7. [DOI: 10.1016/j.exer.2013.09.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 09/11/2013] [Accepted: 09/27/2013] [Indexed: 12/31/2022]
|
25
|
Hsu WH, Lee BH, Hsu YW, Pan TM. Inhibition of Th2 cytokine production in T cells by monascin via PPAR-γ activation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2013; 61:8126-8133. [PMID: 23848565 DOI: 10.1021/jf402373z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Yellow pigment monascin (MS) is a secondary metabolite isolated from Monascus -fermented products and has numerous physiological activities. However, the potential use of MS for immunomodulation remains unclear. We showed that MS and the synthetic peroxisome proliferator-activated receptor (PPAR)-γ ligand rosiglitazone (RG) significantly inhibited the production of Th2 cytokines, including IL-4, IL-5, and IL-13, in PMA/ionomycin-activated mouse EL-4 T cells. Moreover, we showed that this was due to cellular PPAR-γ translocation. These results indicate that MS and RG promote PPAR-γ-DNA interactions and suggest that the regulatory effects of MS and RG on Th2 cytokine production could be abolished with PPAR-γ antagonist treatment. MS and RG also suppressed Th2 transcription factor translocation (e.g., GATA-3 and nuclear factor of activated T cells) by preventing the phosphorylation of protein kinase C and signal transducer and activator of transcription 6.
Collapse
Affiliation(s)
- Wei-Hsuan Hsu
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan
| | | | | | | |
Collapse
|
26
|
Waickman AT, Powell JD. mTOR, metabolism, and the regulation of T-cell differentiation and function. Immunol Rev 2013; 249:43-58. [PMID: 22889214 DOI: 10.1111/j.1600-065x.2012.01152.x] [Citation(s) in RCA: 302] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Upon antigen recognition, naive T cells undergo rapid expansion and activation. The energy requirements for this expansion are formidable, and T-cell activation is accompanied by dramatic changes in cellular metabolism. Furthermore, the outcome of antigen engagement is guided by multiple cues derived from the immune microenvironment. Mammalian target of rapamycin (mTOR) is emerging as a central integrator of these signals playing a critical role in driving T-cell differentiation and function. Indeed, multiple metabolic programs are controlled by mTOR signaling. In this review, we discuss the role of mTOR in regulating metabolism and how these pathways intersect with the ability of mTOR to integrate cues that guide the outcome of T-cell receptor engagement.
Collapse
Affiliation(s)
- Adam T Waickman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | |
Collapse
|
27
|
Relevance of nuclear receptor expression in a Tchreg cell line, HOZOT: RXRα and PPARγ negatively regulate IFN-γ production. RESULTS IN IMMUNOLOGY 2012; 2:158-65. [PMID: 24371580 DOI: 10.1016/j.rinim.2012.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/08/2012] [Accepted: 08/13/2012] [Indexed: 11/24/2022]
Abstract
Nuclear receptors (NRs) have recently received much attention for their newly discovered roles in T cell development, as exemplified by RARα (Treg cells) and RORγt (Th17 cells). In previous studies, we characterized a new type of T cell subset, designated as Tchreg (cytotoxic, helper, and regulatory T) cells, in terms of its cytokine signature. In this study, we investigated the expression and functional relevance of NRs in Tchreg cells by performing mRNA profiling of HOZOT, a cord blood-derived Tchreg cell line. We identified eleven inducible and eight constitutively expressed NRs in HOZOT. Among these NRs, RXRα and PPARγ showed features of signature NRs of Tchreg cells because they were selectively expressed in HOZOT compared with other T cell subsets. These NRs exhibited contrasting expression patterns, as RXRα was independent of anti-CD3/28 antibody stimulation while PPARγ was stimulated-dependent. Upon agonist treatment, both proteins translocated to the nucleus and inhibited IFN-γ production through binding to the promoter region of the IFN-γ gene. These results provide new insight into the roles of RXRα and PPARγ in T cell biology, especially in their biological relevance in Tchreg cells.
Collapse
|
28
|
Borsting E, Cheng VPC, Glass CK, Vallon V, Cunard R. Peroxisome proliferator-activated receptor-γ agonists repress epithelial sodium channel expression in the kidney. Am J Physiol Renal Physiol 2011; 302:F540-51. [PMID: 22169011 DOI: 10.1152/ajprenal.00306.2011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Thiazolidinediones (TZDs), known as peroxisome proliferator-activated receptor (PPAR) agonists, are used to treat type 2 diabetes. However, ∼5% of patients experience the treatment-limiting side effect of edema. Studies have implicated activation of the epithelial sodium channel (ENaC) as a cause of TZD-induced fluid retention, although there have been conflicting reports. The goal of this study was to resolve the role of PPARγ in control of ENaC isoforms in the kidney. Herein, we demonstrate in mice that rosiglitazone (RGZ), a PPARγ ligand, increases body weight and abdominal fat pad fluid content and reduces hematocrit. Seven days of RGZ decreases ENaCα and ENaCβ mRNA and ENaCγ protein expression in the kidney cortex, and acute treatment for 5 h with pioglitazone, another potent TZD, does not increase renal ENaC isoform mRNA or protein expression. Pioglitazone also decreases ENaCα and ENaCγ mRNA expression in a cortical collecting duct cell line. As no direct transcriptional studies had been conducted, we examined the PPARγ-dependent regulation of ENaC. Pioglitazone represses ENaCγ promoter activity, and this repression is partially relieved by inhibition of protein synthesis. Chromatin immunoprecipitation assays revealed that repression is associated with a decrease in histone H4K5 acetylation at the proximal ENaCγ promoter. In summary, TZDs do not increase ENaC mRNA expression in the kidney, and in fact repress the ENaCγ promoter via an indirect transcriptional mechanism.
Collapse
Affiliation(s)
- Emily Borsting
- Research and Medicine Services, Div. of Nephrology, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA
| | | | | | | | | |
Collapse
|
29
|
Abstract
PPARs, most notably PPAR-gamma, play a crucial role in regulating the activation of alveolar macrophages, which in turn occupy a pivotal place in the immune response to pathogens and particulates drawn in with inspired air. In this review, we describe the dual role of the alveolar macrophage as both a first-line defender through its phagocytotic activity and a regulator of the immune response. Depending on its state of activation, the alveolar macrophage may either enhance or suppress different aspects of immune function in the lung. We then review the role of PPAR-gamma and its ligands in deactivating alveolar macrophages-thus limiting the inflammatory response that, if unchecked, could threaten the essential respiratory function of the alveolus-while upregulating the cell's phagocytotic activity. Finally, we examine the role that inadequate or inappropriate PPAR-gamma responses play in specific lung diseases.
Collapse
|
30
|
Esposito E, Cuzzocrea S. Targeting the peroxisome proliferator-activated receptors (PPARs) in spinal cord injury. Expert Opin Ther Targets 2011; 15:943-59. [DOI: 10.1517/14728222.2011.581231] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
31
|
Raman P, Kaplan BLF, Thompson JT, Vanden Heuvel JP, Kaminski NE. 15-Deoxy-delta12,14-prostaglandin J2-glycerol ester, a putative metabolite of 2-arachidonyl glycerol, activates peroxisome proliferator activated receptor gamma. Mol Pharmacol 2011; 80:201-9. [PMID: 21511917 DOI: 10.1124/mol.110.070441] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
2-Arachidonyl glycerol (2-AG) is an endogenous arachidonic acid derivative capable of suppressing interleukin (IL)-2 production by activated T cells. 2-AG-mediated IL-2 suppression is dependent on cyclooxygenase-2 (COX-2) metabolism and peroxisome proliferator activated receptor γ (PPARγ) activation. The objective of the present studies was to examine whether 15-deoxy-Δ(12,14)-PGJ(2)-glycerol ester (15d-PGJ(2)-G), a putative metabolite of 2-AG, can mimic the actions of 2-AG on IL-2 regulation through PPARγ activation. 15d-PGJ(2)-G bound PPARγ-ligand binding domain in a PPARγ competitive binding assay. 15d-PGJ(2)-G treatment activated PPARγ in a reporter assay, and PPARγ activation was attenuated when a PPARγ antagonist, 2-chloro-5-nitro-N-4-pyridinylbenzamide (T0070907), was present. 15d-PGJ(2)-G treatment suppressed IL-2 production by activated Jurkat cells, which was partially attenuated when pretreated with T0070907. Moreover, IL-2 suppression was pronounced when 15d-PGJ(2)-G was present 30 min before or after T-cell activation. Concordant with IL-2 suppression, 15d-PGJ(2)-G treatment decreased nuclear factor of activated T cells (NFAT) transcriptional activity in transiently transfected Jurkat cells. It is noteworthy that T0070907 alone markedly increased NFAT reporter activity, suggesting the existence of endogenous PPARγ activation and modulation of NFAT. Because COX-2 metabolism of 2-AG is important for IL-2 suppression, the effect of 2-AG on COX-2 and PPARγ mRNA expression was investigated. 2-AG treatment decreased the up-regulation of COX-2 mRNA after T-cell activation, which suggests negative feedback limiting COX-2-mediated metabolism of 2-AG. PPARγ mRNA expression was increased upon activation, and 2-AG treatment produced a modest decrease in PPARγ mRNA expression. Collectively, our findings suggest that 15d-PGJ(2)-G activates PPARγ to decrease NFAT transcriptional activity and IL-2 expression in activated T cells.
Collapse
Affiliation(s)
- Priyadarshini Raman
- Department of Pharmacology & Toxicology and the Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824-1317, USA
| | | | | | | | | |
Collapse
|
32
|
Zhao W, Wang L, Zhang M, Wang P, Zhang L, Yuan C, Qi J, Qiao Y, Kuo PC, Gao C. Peroxisome proliferator-activated receptor gamma negatively regulates IFN-beta production in Toll-like receptor (TLR) 3- and TLR4-stimulated macrophages by preventing interferon regulatory factor 3 binding to the IFN-beta promoter. J Biol Chem 2010; 286:5519-28. [PMID: 21148557 DOI: 10.1074/jbc.m110.149823] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Toll-like receptors 3 and 4 utilize adaptor TRIF to activate interferon regulatory factor 3 (IRF3), resulting in IFN-β production to mediate anti-viral and bacterial infection. Peroxisome proliferator-activated receptor (PPAR)-γ is a ligand-activated transcription factor expressed in various immune cells and acts as a transcriptional repressor to inhibit the transcription of many proinflammatory cytokines. But, the function of PPAR-γ in TLR3- and -4-mediated IFN-β production is not well elucidated. Here, we have analyzed the effect of the PPAR-γ agonists on IFN-β production in peritoneal primary macrophages in response to LPS and poly(I:C). PPAR-γ agonists inhibited LPS and poly(I:C)-induced IFN-β transcription and secretion. siRNA knockdown of PPAR-γ expression and transfection of PPAR-γ expression plasmid demonstrated that PPAR-γ agonist inhibits IFN-β production in a PPAR-γ-dependent manner. The ability of the PPAR-γ agonist to inhibit IFN-β production was confirmed in vivo as mice treated with troglitazone exhibited decreased levels of IFN-β upon LPS and poly(I:C) challenge. Chromatin immunoprecipitation (CHIP) assay and electrophoretic mobility shift assay (EMSA) demonstrated that troglitazone treatment impaired IRF3 binding to the IFN-β promoter. Furthermore, troglitazone could inhibit LPS and poly(I:C)-induced STAT1 phosphorylation and subsequent ISRE activation. These results demonstrate that PPAR-γ negatively regulates IFN-β production in TLR3- and 4-stimulated macrophages by preventing IRF3 binding to the IFN-β promoter.
Collapse
Affiliation(s)
- Wei Zhao
- Key Laboratory for Experimental Teratology of the Ministry of Education, Department of Immunology, Shandong University Medical School, Jinan, Shandong 250012, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Heneka MT, O'Banion MK, Terwel D, Kummer MP. Neuroinflammatory processes in Alzheimer's disease. J Neural Transm (Vienna) 2010; 117:919-47. [PMID: 20632195 DOI: 10.1007/s00702-010-0438-z] [Citation(s) in RCA: 323] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 06/16/2010] [Indexed: 12/12/2022]
Abstract
Generation of neurotoxic amyloid beta peptides and their deposition along with neurofibrillary tangle formation represent key pathological hallmarks in Alzheimer's disease (AD). Recent evidence suggests that inflammation may be a third important component which, once initiated in response to neurodegeneration or dysfunction, may actively contribute to disease progression and chronicity. Various neuroinflammatory mediators including complement activators and inhibitors, chemokines, cytokines, radical oxygen species and inflammatory enzyme systems are expressed and released by microglia, astrocytes and neurons in the AD brain. Degeneration of aminergic brain stem nuclei including the locus ceruleus and the nucleus basalis of Meynert may facilitate the occurrence of inflammation in their projection areas given the antiinflammatory and neuroprotective action of their key transmitters norepinephrine and acetylcholine. While inflammation has been thought to arise secondary to degeneration, recent experiments demonstrated that inflammatory mediators may stimulate amyloid precursor protein processing by various means and therefore can establish a vicious cycle. Despite the fact that some aspects of inflammation may even be protective for bystander neurons, antiinflammatory treatment strategies should therefore be considered. Non-steroidal anti-inflammatory drugs have been shown to reduce the risk and delay the onset to develop AD. While, the precise molecular mechanism underlying this effect is still unknown, a number of possible mechanisms including cyclooxygenase 2 or gamma-secretase inhibition and activation of the peroxisome proliferator activated receptor gamma may alone or, more likely, in concert account for the epidemiologically observed protection.
Collapse
Affiliation(s)
- Michael T Heneka
- Department of Neurology, Clinical Neurosciences, University of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany.
| | | | | | | |
Collapse
|
34
|
Kiss E, Popovic ZV, Bedke J, Adams J, Bonrouhi M, Babelova A, Schmidt C, Edenhofer F, Zschiedrich I, Domhan S, Abdollahi A, Schäfer L, Gretz N, Porubsky S, Gröne HJ. Peroxisome proliferator-activated receptor (PPAR)gamma can inhibit chronic renal allograft damage. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2150-62. [PMID: 20363918 PMCID: PMC2861081 DOI: 10.2353/ajpath.2010.090370] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/06/2010] [Indexed: 12/19/2022]
Abstract
Chronic inflammation and fibrosis are the leading causes of chronic allograft failure. The nuclear receptor peroxisome proliferator-activated receptor (PPAR)gamma is a transcription factor known to have antidiabetogenic and immune effects, and PPARgamma forms obligate heterodimers with the retinoid X receptor (RXR). We have reported that a retinoic acid (RAR)/RXR-agonist can potently influence the course of renal chronic allograft dysfunction. In this study, in a Fischer to Lewis rat renal transplantation model, administration of the PPARgamma-agonist, rosiglitazone, independent of dose (3 or 30 mg/kgBW/day), lowered serum creatinine, albuminuria, and chronic allograft damage with a chronic vascular damage score as follows: 35.0 +/- 5.8 (controls) vs. 8.1 +/- 2.4 (low dose-Rosi; P < 0.05); chronic tubulointerstitial damage score: 13.6 +/- 1.8 (controls) vs. 2.6 +/- 0.4 (low dose-Rosi; P < 0.01). The deposition of extracellular matrix proteins (collagen, fibronectin, decorin) was strikingly lower. The expression of transforming growth factor-beta1 was inhibited, whereas that of bone morphogenic protein-7 (BMP-7) was increased. Intragraft mononuclear cells and activated fibroblast numbers were reduced by 50%. In addition, the migratory and proliferative activity of these cells was significantly inhibited in vitro. PPARgamma activation diminished the number of cells expressing the proinflammatory and fibrogenic proteoglycan biglycan. In macrophages its secretion was blocked by rosiglitazone in a predominantly PPARgamma-dependent manner. The combination of PPARgamma- and RAR/RXR-agonists resulted in additive effects in the inhibition of fibrosis. In summary, PPARgamma activation was potently immunosuppressive and antifibrotic in kidney allografts, and these effects were enhanced by a RAR/RXR-agonist.
Collapse
Affiliation(s)
- Eva Kiss
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Yang KM, Ntrivalas E, Cho HJ, Kim NY, Beaman K, Gilman-Sachs A, Kwak-Kim J. ORIGINAL ARTICLE: Women with Multiple Implantation Failures and Recurrent Pregnancy Losses have Increased Peripheral Blood T Cell Activation. Am J Reprod Immunol 2010; 63:370-8. [DOI: 10.1111/j.1600-0897.2010.00811.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
36
|
Won HY, Min HJ, Ahn JH, Yoo SE, Bae MA, Hong JH, Hwang ES. Anti-allergic function and regulatory mechanisms of KR62980 in allergen-induced airway inflammation. Biochem Pharmacol 2009; 79:888-96. [PMID: 19896472 DOI: 10.1016/j.bcp.2009.10.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 10/27/2009] [Accepted: 10/27/2009] [Indexed: 11/16/2022]
Abstract
The ligand-activated transcription factor, peroxisome proliferator-activated receptor (PPAR)gamma, and its ligands inhibit pro-inflammatory cytokine production by immune cells, thus exerting anti-inflammatory activity. As a non-thiazolidinedione PPARgamma ligand, KR62980 has anti-diabetic and anti-adipogenic activities, but its anti-inflammatory function has yet to be characterized. In this study, we investigated the functions and mechanisms of KR62980 in the activation and differentiation of CD4+ T helper (Th) cells by comparing its effects with those of a thiazolidinedione PPARgamma ligand, rosiglitazone. KR62980 dose-dependently and significantly suppressed TCR-triggered Th cell proliferation by suppressing IL-2/IL-2Ralpha-mediated signaling. Both KR62980 and rosiglitazone suppressed IFNgamma production in a dose-dependent manner, whereas IL-4 gene expression was specifically suppressed by only KR62980. In addition, sustained KR62980 treatment diminished Th2 cytokine production by inhibiting c-Maf expression. In vivo administration of KR62980 in a model of allergic asthma significantly attenuated eotaxin-induced eosinophil infiltration, allergic cytokine production and collagen deposition in the lung. KR62980 also decreased goblet cell hyperplasia in the airway and mucous cell metaplasia in nasal epithelium, concurrent with decreases of allergic Th2 cytokines and IL-17 in the draining lymph node. In conclusion, a novel PPARgamma ligand, KR62980, suppresses in vitro Th2 cell differentiation and attenuates in vivo OVA-induced airway inflammation, suggesting a beneficial role for KR62980 in the treatment of allergic asthma and allergic rhinitis.
Collapse
Affiliation(s)
- Hee Yeon Won
- College of Pharmacy and Division of Life and Pharmaceutical Sciences and Center for Cell Signaling & Drug Discovery Research, Ewha Womans University, 11-1 Daehyun-Dong, Sudaemun-Ku, Seoul 120-750, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
37
|
Spelman K, Iiams-Hauser K, Cech NB, Taylor EW, Smirnoff N, Wenner CA. Role for PPARγ in IL-2 inhibition in T cells by Echinacea-derived undeca-2E-ene-8,10-diynoic acid isobutylamide. Int Immunopharmacol 2009; 9:1260-4. [DOI: 10.1016/j.intimp.2009.08.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 08/16/2009] [Accepted: 08/17/2009] [Indexed: 12/15/2022]
|
38
|
Klotz L, Hucke S, Thimm D, Classen S, Gaarz A, Schultze J, Edenhofer F, Kurts C, Klockgether T, Limmer A, Knolle P, Burgdorf S. Increased antigen cross-presentation but impaired cross-priming after activation of peroxisome proliferator-activated receptor gamma is mediated by up-regulation of B7H1. THE JOURNAL OF IMMUNOLOGY 2009; 183:129-36. [PMID: 19535643 DOI: 10.4049/jimmunol.0804260] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Dendritic cells are able to take up exogenous Ags and present Ag-derived peptides on MHC class I molecules, a process termed cross-presentation. The mannose receptor (MR), an endocytic receptor expressed on a variety of APCs, has been demonstrated to target soluble Ags exclusively toward cross-presentation. In this study, we investigated the role of the murine nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma), a ligand-activated transcription factor with immunomodulatory properties, in MR-mediated endocytosis and cross-presentation of the model Ag OVA. We could demonstrate both in vitro and in vivo that activation of PPARgamma resulted in increased MR expression, which in consequence led to enhanced MR-mediated endocytosis and elevated cross-presentation of soluble OVA. Concomitantly, activation of PPARgamma in dendritic cells induced up-regulation of the coinhibitory molecule B7H1, which, despite enhanced cross-presentation, caused an impaired activation of naive OVA-specific CD8(+) T cells and the induction of T cell tolerance. These data provide a mechanistic basis for the immunomodulatory action of PPARgamma which might open new possibilities in the development of therapeutic approaches aimed at the control of excessive immune responses, e.g., in T cell-mediated autoimmunity.
Collapse
Affiliation(s)
- Luisa Klotz
- Institutes of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zhou Q, Liao JK. Statins and cardiovascular diseases: from cholesterol lowering to pleiotropy. Curr Pharm Des 2009; 15:467-78. [PMID: 19199975 DOI: 10.2174/138161209787315684] [Citation(s) in RCA: 186] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Statins are 3-hydroxy-3-methyglutaryl coenzyme A (HMG-CoA) reductase inhibitors, which are prescribed extensively for cholesterol lowering in the primary and secondary prevention of cardiovascular disease. Recent compelling evidence suggests that the beneficial effects of statins may not only be due to their cholesterol lowering effects, but also, to their cholesterol-independent or pleiotropic effects. Through these so-called pleiotropic effects, statins are directly involved in restoring or improving endothelial function, attenuating vascular remodeling, inhibiting vascular inflammatory response, and perhaps, stabilizing atherosclerotic plaques. These cholesterol-independent effects of statins are predominantly due to their ability to inhibit isoprenoid synthesis, the products of which are important lipid attachments for intracellular signaling molecules, such as Rho, Rac and Cdc42. In particular, inhibition of Rho and its downstream target, Rho-associated coiled-coil containing protein kinase (ROCK), has emerged as the principle mechanisms underlying the pleiotropic effects of statins. This review provides an update of statin-mediated vascular effects beyond cholesterol lowering and highlights recent findings from bench to bedside to support the concept of statin pleiotropy.
Collapse
Affiliation(s)
- Qian Zhou
- Vascular Medicine Research Unit, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02139, USA
| | | |
Collapse
|
40
|
Malur A, Mccoy AJ, Arce S, Barna BP, Kavuru MS, Malur AG, Thomassen MJ. Deletion of PPARγ in Alveolar Macrophages Is Associated with a Th-1 Pulmonary Inflammatory Response. THE JOURNAL OF IMMUNOLOGY 2009; 182:5816-22. [DOI: 10.4049/jimmunol.0803504] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
41
|
Gocke AR, Hussain RZ, Yang Y, Peng H, Weiner J, Ben LH, Drew PD, Stuve O, Lovett-Racke AE, Racke MK. Transcriptional modulation of the immune response by peroxisome proliferator-activated receptor-{alpha} agonists in autoimmune disease. THE JOURNAL OF IMMUNOLOGY 2009; 182:4479-87. [PMID: 19299749 DOI: 10.4049/jimmunol.0713927] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Peroxisome proliferator-activated receptor-alpha (PPARalpha) agonists have been shown to have a therapeutic benefit in experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis (MS). In this study, we investigated the mechanism by which the PPARalpha agonist gemfibrozil induces immune deviation and protects mice from EAE. We demonstrated that treatment with gemfibrozil increases expression of the Th2 transcription factor GATA-3 and decreases expression of the Th1 transcription factor T-bet in vitro and directly ex vivo. These changes correlated with an increase in nuclear PPARalpha expression. Moreover, the protective effects of PPARalpha agonists in EAE were shown to be partially dependent on IL-4 and to occur in a receptor-dependent manner. PPARalpha was demonstrated, for the first time, to regulate the IL-4 and IL-5 genes and to bind the IL-4 promoter in the presence of steroid receptor coactivator-1, indicating that PPARalpha can directly transactivate the IL-4 gene. Finally, therapeutic administration of PPARalpha agonists ameliorated clinically established EAE, suggesting that PPARalpha agonists may provide a treatment option for immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Anne R Gocke
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, 75390, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Peroxisome proliferator-activated receptors in HCV-related infection. PPAR Res 2009; 2009:357204. [PMID: 19343188 PMCID: PMC2662434 DOI: 10.1155/2009/357204] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2008] [Accepted: 01/06/2009] [Indexed: 12/31/2022] Open
Abstract
The topic of peroxisome proliferator-activated receptors has been developed in the field of hepatology allowing envisaging therapeutic strategies for the most frequent chronic liver diseases such as chronic infection with hepatitis C virus (HCV). PPARs contribute to wide physiological processes within the liver such as lipid/glucid metabolisms, inflammatory response, cell differentiation, and cell cycle. In vitro experiments and animal studies showed that PPARα discloses anti-inflammatory property, and PPARγ discloses anti-inflammatory, antifibrogenic, and antiproliferative properties in the liver. Experimental and human studies showed impaired PPARs expression and function during HCV infection. The available nonhepatotoxic agonists of PPARs may constitute a progress in the therapeutic management of patients chronically infected with HCV.
Collapse
|
43
|
Repression of interferon-gamma expression in T cells by Prospero-related homeobox protein. Cell Res 2009; 18:911-20. [PMID: 19160541 DOI: 10.1038/cr.2008.275] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Interferon-gamma (IFN-gamma) is a major proinflammatory effector and regulatory cytokine produced by activated T cells and NK cells. IFN-gamma has been shown to play pivotal roles in fundamental immunological processes such as inflammatory reactions, cell-mediated immunity and autoimmunity. A variety of human disorders have now been linked to irregular IFN-gamma expression. In order to achieve proper IFN-gamma-mediated immunological effects, IFN-gamma expression in T cells is subject to both positive and negative regulation. In this study, we report for the first time the negative regulation of IFN-gamma expression by Prospero-related Homeobox (Prox1). In Jurkat T cells and primary human CD4+ T cells, Prox1 expression decreases quickly upon T cell activation, concurrent with a dramatic increase in IFN-gamma expression. Reporter analysis and chromatin immunoprecipitation (ChIP) revealed that Prox1 associates with and inhibits the transcription activity of IFN-,gammapromoter in activated Jurkat T cells. Co-immunoprecipitation and GST pull-down assay demonstrated a direct binding between Prox1 and the nuclear receptor peroxisome proliferator-activated receptor gamma (PPPARgamma, which is also an IFN-gamma repressor in T cells. By introducing deletions and mutations into Prox1, we show that the repression of IFN-gamma promoter by Prox1 is largely dependent upon the physical interaction between Prox1 and PPPARgamma Furthermore, PPPARgammaantagonist treatment removes Prox1 from IFN-gamma promoter and attenuates repression of IFN-gamma expression by Prox1. These findings establish Prox1 as a new negative regulator of IFN-gamma expression in T cells and will aid in the understanding of IFN-gamma transcription regulation mechanisms.
Collapse
|
44
|
Jeon EJ, Lee SK, Park YS, Kim DH, Yum JH, Park CS. The effects of Peroxisome Proliferator-Activated Receptor-γ agonist on a murine model of experimental allergic rhinitis. Otolaryngol Head Neck Surg 2008; 139:124-30. [DOI: 10.1016/j.otohns.2008.03.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Revised: 03/21/2008] [Accepted: 03/27/2008] [Indexed: 11/29/2022]
Abstract
Objectives To investigate the effect of rosiglitazone, a synthetic selective peroxisome proliferator-activated receptor (PPAR)-γ agonist, for cytokine production and PPAR-γ expression in nasal mucosa. Methods Mice in allergic rhinitis group received ovalbumin sensitization followed by ovalbumin intranasal challenge. Mice in the rosiglitazone group received rosiglitazone treatment additionally. Various allergic responses were then assessed. Results The frequency of nasal rubs and ovalbumin-specific immunoglobulin E decreased significantly in the rosiglitazone group compared with the allergic rhinitis group. The rosiglitazone group also showed that inflammation was markedly reduced by rosiglitazone administration. Immunohistochemistry showed that PPAR-γ protein expression in nasal mucosa was enhanced in the allergic rhinitis group and the rosiglitazone group compared with control mice. Conclusion PPAR-γ activation with rosiglitazone effectively inhibited allergic symptom development, nasal mucosal inflammation, and production of ovoalbumin-specific immunoglobulin E and Th2-type cytokine. Our results provide evidence of the therapeutic potential of PPAR-γ agonist for the treatment of allergic rhinitis.
Collapse
Affiliation(s)
- Eun-Ju Jeon
- Department of Otolaryngology-HNS, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung Kyun Lee
- Department of Otolaryngology-HNS, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yong-Soo Park
- Department of Otolaryngology-HNS, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Dong-Hyun Kim
- Department of Otolaryngology-HNS, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jin Ho Yum
- Department of Otolaryngology-HNS, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chan-Soon Park
- Department of Otolaryngology-HNS, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
45
|
Fang X, Zhang L, Feng Y, Zhao Y, Dai J. Immunotoxic effects of perfluorononanoic acid on BALB/c mice. Toxicol Sci 2008; 105:312-21. [PMID: 18583369 DOI: 10.1093/toxsci/kfn127] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The effects of perfluorononanoic acid (PFNA) on the immune system and its mechanism of action in mice have not been elucidated. Thus, BALB/c mice were exposed to the PFNA (0, 1, 3, or 5 mg/kg/day) for fourteen days. Exposure to PFNA led to a decrease in body weight and in the weight of the lymphoid organs. Cell cycle arrest and apoptosis were observed in the spleen and thymus following PFNA exposure. In the thymus, PFNA mostly modulated CD4+CD8+ thymocytes, whereas the F4/80+, CD11c+, and CD49b+ cells were major targets in the spleen. Although concanavalin A-induced T lymphocyte blastogenesis was not altered by PFNA, production of interleukin (IL)-4 and interferon-gamma by splenic lymphocytes was remarkably impaired. The levels of cortisol and adrenocorticotrophic hormone in sera were increased; however, the expression of glucocorticoid receptor in the thymus was unchanged. In addition, expression of the peroxisome proliferator-activated receptors (PPAR-alpha and PPAR-gamma) and IL-1beta were upregulated significantly in the thymus at a dose of 1 mg PFNA/kg/day. No significant changes in expression of the inhibitory protein IkappaBalpha and IkappaBalpha kinase were observed. Together, these results suggest that PFNA exerts toxic effects on lymphoid organs and T cell and innate immune cell homeostasis in mice and that these effects may result from the activation of PPAR-alpha, PPAR-gamma, and the hypothalamic-pituitary-adrenal axis. Interestingly, at the transcriptional level, the nuclear factor-kappa B signaling pathway appears to be uninvolved in the immunotoxic potential of PFNA.
Collapse
Affiliation(s)
- Xuemei Fang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100190, China
| | | | | | | | | |
Collapse
|
46
|
Dharancy S, Louvet A, Hollebecque A, Desreumaux P, Mathurin P, Dubuquoy L. [Nuclear receptor PPAR and hepatology: pathophysiological and therapeutical aspects]. ACTA ACUST UNITED AC 2008; 32:339-50. [PMID: 18396382 DOI: 10.1016/j.gcb.2008.01.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2007] [Revised: 12/18/2007] [Accepted: 01/05/2008] [Indexed: 01/08/2023]
Abstract
In last few years, the topic of nuclear receptor has been developed in the field of hepatology allowing envisaging therapeutic strategies for the most frequent chronic liver diseases. Peroxysome proliferator-activated receptors (PPAR) contribute to wide physiological processes within the liver such as lipid/glucid metabolisms, inflammatory response, cell differenciation and cell cycle. In vitro experiments and animal studies showed that PPARalpha discloses anti-inflammatory property and PPARgamma discloses anti-inflammatory, antifibrogenic and antiproliferative properties in the liver. Main available agonists are fibrates (PPARalpha) used for 20 years in cases of lipid metabolism abnormalities and glitazones (PPARgamma) used since 2000 for type 2 diabetes. In terms of therapy, animal studies and human trials have been conducted in steatopathies. However, clinicians have to be aware of potential specific side effects related to glitazones especially on cardiovascular system.
Collapse
Affiliation(s)
- S Dharancy
- Inserm U795, Boulevard du Professeur-Jules-Leclercq, 59037 Lille, France.
| | | | | | | | | | | |
Collapse
|
47
|
Narala VR, Ranga R, Smith MR, Berlin AA, Standiford TJ, Lukacs NW, Reddy RC. Pioglitazone is as effective as dexamethasone in a cockroach allergen-induced murine model of asthma. Respir Res 2007; 8:90. [PMID: 18053220 PMCID: PMC2231357 DOI: 10.1186/1465-9921-8-90] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Accepted: 12/04/2007] [Indexed: 01/17/2023] Open
Abstract
Background While glucocorticoids are currently the most effective therapy for asthma, associated side effects limit enthusiasm for their use. Peroxisome proliferator-activated receptor-γ (PPAR-γ) activators include the synthetic thiazolidinediones (TZDs) which exhibit anti-inflammatory effects that suggest usefulness in diseases such as asthma. How the ability of TZDs to modulate the asthmatic response compares to that of glucocorticoids remains unclear, however, because these two nuclear receptor agonists have never been studied concurrently. Additionally, effects of PPAR-γ agonists have never been examined in a model involving an allergen commonly associated with human asthma. Methods We compared the effectiveness of the PPAR-γ agonist pioglitazone (PIO) to the established effectiveness of a glucocorticoid receptor agonist, dexamethasone (DEX), in a murine model of asthma induced by cockroach allergen (CRA). After sensitization to CRA and airway localization by intranasal instillation of the allergen, Balb/c mice were challenged twice at 48-h intervals with intratracheal CRA. Either PIO (25 mg/kg/d), DEX (1 mg/kg/d), or vehicle was administered throughout the period of airway CRA exposure. Results PIO and DEX demonstrated similar abilities to reduce airway hyperresponsiveness, pulmonary recruitment of inflammatory cells, serum IgE, and lung levels of IL-4, IL-5, TNF-α, TGF-β, RANTES, eotaxin, MIP3-α, Gob-5, and Muc5-ac. Likewise, intratracheal administration of an adenovirus containing a constitutively active PPAR-γ expression construct blocked CRA induction of Gob-5 and Muc5-ac. Conclusion Given the potent effectiveness shown by PIO, we conclude that PPAR-γ agonists deserve investigation as potential therapies for human asthma.
Collapse
Affiliation(s)
- Venkata R Narala
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109-2200, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Pascual G, Ricote M, Hevener AL. Macrophage peroxisome proliferator activated receptor γ as a therapeutic target to combat Type 2 diabetes. Expert Opin Ther Targets 2007; 11:1503-20. [DOI: 10.1517/14728222.11.11.1503] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
49
|
Klotz L, Diehl L, Dani I, Neumann H, von Oppen N, Dolf A, Endl E, Klockgether T, Engelhardt B, Knolle P. Brain endothelial PPARγ controls inflammation-induced CD4+ T cell adhesion and transmigration in vitro. J Neuroimmunol 2007; 190:34-43. [PMID: 17719655 DOI: 10.1016/j.jneuroim.2007.07.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2007] [Revised: 07/06/2007] [Accepted: 07/25/2007] [Indexed: 11/19/2022]
Abstract
An important step in the pathogenesis of multiple sclerosis is adhesion and transmigration of encephalitogenic T cells across brain endothelial cells (EC) which strongly relies on interaction with EC-expressed adhesion molecules. We provide molecular evidence that the transcription factor peroxisome proliferator-activated receptor gamma (PPARgamma) is a negative regulator of brain EC inflammation. The PPARgamma agonist pioglitazone reduces transendothelial migration of encephalitogenic T cells across TNFalpha-stimulated brain EC. This effect is clearly PPARgamma mediated, as lentiviral PPARgamma overexpression in brain EC results in selective abrogation of inflammation-induced ICAM-1 and VCAM-1 upregulation and subsequent adhesion and transmigration of T cells. We therefore propose that PPARgamma in brain EC may be exploited to target detrimental EC-T cell interactions under inflammatory conditions.
Collapse
Affiliation(s)
- Luisa Klotz
- Institute of Molecular Medicine and Experimental Immunology, University of Bonn, Sigmund-Freud-Strasse 25, Bonn, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Fionda C, Nappi F, Piccoli M, Frati L, Santoni A, Cippitelli M. Inhibition of trail gene expression by cyclopentenonic prostaglandin 15-deoxy-delta12,14-prostaglandin J2 in T lymphocytes. Mol Pharmacol 2007; 72:1246-57. [PMID: 17673570 DOI: 10.1124/mol.107.038042] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
15-Deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)) is a cyclopentenonic prostaglandin endowed with powerful anti-inflammatory activities, as shown in animal models of inflammatory/autoimmune diseases, where pharmacological administration of this prostanoid can ameliorate inflammation and local tissue damage via activation of the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma) and/or covalent modifications of cellular proteins. Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily expressed in most of the cells, including those of immune system such as T lymphocytes, in which it is up-regulated upon antigen-specific stimulation. This cytokine plays an important role in regulating various physiological and immunopathological processes, such as immunosurveillance of tumors and tissue destruction associated with different inflammatory and autoimmune diseases. Here, we demonstrate that 15d-PGJ(2) inhibits trail mRNA and protein expression by down-regulating the activity of its promoter in human T lymphocytes. Our data indicate that both the chemically reactive cyclopentenone moiety of 15d-PGJ(2) and the activation of PPARgamma may be involved in this repressive mechanism. We identified nuclear factor kappaB (NF-kappaB) as a direct target of the prostanoid. 15d-PGJ(2) significantly decreases the expression and/or DNA binding of c-rel, RelA, and p50 transcription factors to the NF-kappaB1 site of trail promoter. Moreover, 15d-PGJ(2)-mediated activation of the transcription factor heat shock factor-1 may contribute to inhibit trail promoter activity in transfected Jurkat T cells. These results suggest that modulation of TRAIL gene expression by 15d-PGJ(2) in T cells may provide a novel pharmacological tool to modify the onset and the progression of specific autoimmune and inflammatory disorders.
Collapse
Affiliation(s)
- Cinzia Fionda
- Department of Experimental Medicine, Istituto Pasteur-Fondazione Cenci Bolognetti, University La Sapienza, Viale Regina Elena 324, 00161, Rome, Italy
| | | | | | | | | | | |
Collapse
|