1
|
Back P, Yu M, Modaresahmadi S, Hajimirzaei S, Zhang Q, Islam MR, Schwendeman AA, La-Beck NM. Immune Implications of Cholesterol-Containing Lipid Nanoparticles. ACS NANO 2024; 18:28480-28501. [PMID: 39388645 PMCID: PMC11505898 DOI: 10.1021/acsnano.4c06369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024]
Abstract
The majority of clinically approved nanoparticle-mediated therapeutics are lipid nanoparticles (LNPs), and most of these LNPs are liposomes containing cholesterol. LNP formulations significantly alter the drug pharmacokinetics (PK) due to the propensity of nanoparticles for uptake by macrophages. In addition to readily engulfing LNPs, the high expression of cholesterol hydroxylases and reactive oxygen species (ROS) in macrophages suggests that they will readily produce oxysterols from LNP-associated cholesterol. Oxysterols are a heterogeneous group of cholesterol oxidation products that have potent immune modulatory effects. Oxysterols are implicated in the pathogenesis of atherosclerosis and certain malignancies; they have also been found in commercial liposome preparations. Yet, the in vivo metabolic fate of LNP-associated cholesterol remains unclear. We review herein the mechanisms of cellular uptake, trafficking, metabolism, and immune modulation of endogenous nanometer-sized cholesterol particles (i.e., lipoproteins) that are also relevant for cholesterol-containing nanoparticles. We believe that it would be imperative to better understand the in vivo metabolic fate of LNP-associated cholesterol and the immune implications for LNP-therapeutics. We highlight critical knowledge gaps that we believe need to be addressed in order to develop safer and more efficacious lipid nanoparticle delivery systems.
Collapse
Affiliation(s)
- Patricia
Ines Back
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Minzhi Yu
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, Michigan 48109, United States
| | - Shadan Modaresahmadi
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Sahelosadat Hajimirzaei
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Qisheng Zhang
- Division
of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Md Rakibul Islam
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
| | - Anna A. Schwendeman
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, Michigan 48109, United States
- Biointerfaces
Institute, University of Michigan, North
Campus Research Complex, 2800 Plymouth Road, Ann Arbor, Michigan 48109, United States
| | - Ninh M. La-Beck
- Department
of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of
Pharmacy, Texas Tech University Health Sciences
Center, Abilene, Texas 79601, United States
- Department
of Pharmacy Practice, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, Texas 79601, United States
| |
Collapse
|
2
|
Soni N, Bissa B. Exosomes, Circadian Rhythms, and Cancer Precision Medicine: New Frontiers. Biochimie 2024:S0300-9084(24)00169-X. [PMID: 39032591 DOI: 10.1016/j.biochi.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 07/23/2024]
Abstract
"The environment shapes people's actions," a well-known proverb, strongly dictates that a change in our way of life changes our behavior. Circadian rhythms have been identified as a mechanism for maintaining homeostasis in the body, which, if disrupted by sleeping patterns, could result in significant metabolic alterations that adversely affect our health. The changes induced by circadian rhythm alter the secretion and cargo selection in exosomes which are nanovesicles important for intercellular communication. Exosomes were formerly known as "junk particles" but are now recognized as miniature copies of a cell's genetic material. Dysregulation of circadian rhythm has shown that it changes the gene expression of a cell to some extent and significantly alters the exosomal release. Meanwhile, cells secrete exosomes continuously to align the rhythmicity of the biological clock. In this study, we integrate circadian rhythms and exosomes with precision medicines to find better approaches to early diagnosis and treatment of disease.
Collapse
Affiliation(s)
- Naveen Soni
- Dept. of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Bhawana Bissa
- Dept. of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India.
| |
Collapse
|
3
|
Wang MH, Liu X, Wang Q, Zhang HW. A circadian rhythm-related gene signature for prognosis, invasion and immune microenvironment of breast cancer. Front Genet 2023; 13:1104338. [PMID: 36685904 PMCID: PMC9849377 DOI: 10.3389/fgene.2022.1104338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/19/2022] [Indexed: 01/06/2023] Open
Abstract
Background: Circadian dysregulation is linked to the onset and progression of cancer, but current knowledge of the role of circadian rhythm-related genes (CRRGs) in breast cancer (BC) is limited and incomplete. The purpose of this study was to investigate the potential role and immune-related prognostic significance of CRRGs in BC. Methods: The Cancer Genome Atlas breast cancer (TCGA-BRCA) genetic data were combined with 1369 CRRGs to create a model of BC prognosis-related CRRGs. To validate the model's predictive power in TCGA and other external datasets, the Kaplan-Meier survival curve and receptor operation characteristic curve were plotted. The relationship between CRRGs model and gene enrichment pathways, immune cell infiltration, and differences in patient response to immune checkpoint inhibitors (ICIs) therapy was then discussed. Results: A CRRG-based eighteen-gene model was developed that accurately predicted the survival time of BC patients. Based on this model, BC patients can be classified as high or low risk. The high-risk group has negative immune cell infiltration (such as macrophages M0 and M2) and a poor therapeutic response to ICIs due to lower immune checkpoint gene expression. Furthermore, TCF7 and IFNG were found to be strongly associated with immune checkpoints in CRRGs model. Conclusion: The 18 CRRGs may be useful in assessing the prognosis of BC patients, studying immune infiltration, and developing more effective immunotherapy strategies.
Collapse
Affiliation(s)
- Mei-Huan Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiao Liu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qian Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China,Department of Ultrasound, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China,*Correspondence: Qian Wang, ; Hua-Wei Zhang,
| | - Hua-Wei Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China,Department of Ultrasound, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China,*Correspondence: Qian Wang, ; Hua-Wei Zhang,
| |
Collapse
|
4
|
Ma L, Nelson ER. Oxysterols and nuclear receptors. Mol Cell Endocrinol 2019; 484:42-51. [PMID: 30660701 DOI: 10.1016/j.mce.2019.01.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/08/2019] [Accepted: 01/16/2019] [Indexed: 12/11/2022]
Abstract
Oxysterols are derivatives of cholesterol and an important regulator of cholesterol metabolism, in part due to their role as ligands for nuclear receptors, such as the liver X receptors. Oxysterols are also known to be ligands for the RAR-related orphan receptors, involved in normal T cell differentiation. However, increasing evidence supports a role for oxysterols in the progression of several diseases. Here, we review recent developments in oxysterol research, highlighting the biological functions that oxysterols exert through their target nuclear receptors: the liver X receptors, estrogen receptors, RAR-related orphan receptors and the glucocorticoid receptor. We also bring the regulation of the immune system into the context of interaction between oxysterols and nuclear receptors, discussing the effect of such interaction on the pro-inflammatory function of macrophages and the development of T cells. Finally, we examine the impact that oxysterols have on various disease models, including cancer, Alzheimer's disease and atherosclerosis, stressing the role of nuclear receptors if previously identified. This review underscores the need to consider the multifaceted roles of oxysterols in terms of multiple receptor engagements and selective modulation of these receptors.
Collapse
Affiliation(s)
- Liqian Ma
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States; University of Illinois Cancer Center, Chicago, IL, United States; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Carl R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People Theme, University of Illinois at Urbana Champaign, Urbana, IL, United States; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, IL, United States.
| |
Collapse
|
5
|
He S, Nelson ER. 27-Hydroxycholesterol, an endogenous selective estrogen receptor modulator. Maturitas 2017; 104:29-35. [PMID: 28923174 DOI: 10.1016/j.maturitas.2017.07.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 07/26/2017] [Accepted: 07/28/2017] [Indexed: 12/11/2022]
Abstract
Estrogen receptors (ERs) mediate the actions of the steroidal estrogens, and are important for the regulation of several physiological and pathophysiological processes, including reproduction, bone physiology, cardiovascular physiology and breast cancer. The unique pharmacology of the ERs allows for certain ligands, such as tamoxifen, to elicit tissue- and context-specific responses, ligands now referred to as selective estrogen receptor modulators (SERMs). Recently, the cholesterol metabolite 27-hydroxychoelsterol (27HC) has been defined as an endogenous SERM, with activities in atherosclerosis, osteoporosis, breast and prostate cancers, and neural degenerative diseases. Since 27HC concentrations closely mirror those of cholesterol, it is possible that 27HC mediates many of the biological effects of cholesterol. This paper provides an overview of ER pharmacology and summarizes the work to date implicating 27HC in various diseases. Wherever possible, we highlight clinical data in support of a role for 27HC in the diseases discussed.
Collapse
Affiliation(s)
- Sisi He
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; University of Illinois Cancer Center, Chicago, IL, USA; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
6
|
Umetani M. Re-adopting classical nuclear receptors by cholesterol metabolites. J Steroid Biochem Mol Biol 2016; 157:20-6. [PMID: 26563834 PMCID: PMC4724260 DOI: 10.1016/j.jsbmb.2015.11.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 07/10/2015] [Accepted: 11/04/2015] [Indexed: 12/22/2022]
Abstract
Since the first cloning of the human estrogen receptor (ER) α in 1986 and the subsequent cloning of human ERβ, there has been extensive investigation of the role of estrogen/ER. Estrogens/ER play important roles not only in sexual development and reproduction but also in a variety of other functions in multiple tissues. Selective Estrogen Receptor Modulators (SERMs) are ER lignds that act as agonists or antagonists depending on the target genes and tissues, and until recently, only synthetic SERMs have been recognized. However, the discovery of the first endogenous SERM, 27-hydroxycholesterol (27HC), opened a new dimension of ER action in health and disease. In addition to the identification of 27HC as a SERM, oxysterols have been recently demonstrated as indirect modulators of ER through interaction with the nuclear receptor Liver X Receptor (LXR) β. In this review, the recent progress on these novel roles of oxysterols in ER modulation is summarized.
Collapse
Affiliation(s)
- Michihisa Umetani
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, 3517 Cullen Blvd, SERC 545, Houston, TX 77204-5056, USA.
| |
Collapse
|
7
|
Nelson ER, Chang CY, McDonnell DP. Cholesterol and breast cancer pathophysiology. Trends Endocrinol Metab 2014; 25:649-55. [PMID: 25458418 PMCID: PMC4268141 DOI: 10.1016/j.tem.2014.10.001] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 09/30/2014] [Accepted: 10/07/2014] [Indexed: 01/05/2023]
Abstract
Cholesterol is a risk factor for breast cancer although the mechanisms by which this occurs are not well understood. One hypothesis is that dyslipidemia results in increased cholesterol content in cell membranes, thus impacting upon membrane fluidity and subsequent signaling. In addition, studies demonstrate that the metabolite, 27-hydroxycholesterol (27HC), can function as an estrogen, increasing the proliferation of estrogen receptor (ER)-positive breast cancer cells. This was unexpected because 27HC and other oxysterols activate the liver X receptors (LXR), resulting in a reduction of intracellular cholesterol. Resolution of this paradox will require dissection of the molecular mechanisms by which ER and LXR converge in breast cancer cells. Regardless, the observation that 27HC influences breast cancer provides a rationale for strategies that target cholesterol metabolism.
Collapse
Affiliation(s)
- Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, IL 61801, USA
| | - Ching-yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
8
|
Lee WR, Ishikawa T, Umetani M. The interaction between metabolism, cancer and cardiovascular disease, connected by 27-hydroxycholesterol. ACTA ACUST UNITED AC 2014; 9:617-624. [PMID: 25632306 DOI: 10.2217/clp.14.53] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oxysterols are metabolites of cholesterol that are produced in liver and other peripheral tissues as a means to eliminate cholesterol to bile acid. Recent studies have revealed that the most abundant circulating oxysterol 27-hydroxycholesterol (27HC) is the first identified endogenous selective estrogen receptor modulator. 27HC levels correlate well with that of cholesterol, and also rise progressively with age. 27HC affects estrogen receptor function by the antagonism of estrogen action and also by the direct modulation of the receptor function, and similar to estrogen/estrogen receptors, 27HC has many actions in various tissues. This review article introduces the recent progress in the understanding of the role of 27HC in breast cancer and cardiovascular dysfunction.
Collapse
Affiliation(s)
- Wan-Ru Lee
- Division of Pulmonary & Vascular Biology, Departments of Pediatrics & Pharmacology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Tomonori Ishikawa
- Division of Pulmonary & Vascular Biology, Departments of Pediatrics & Pharmacology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA ; Comprehensive Reproductive Medicine, Graduate School of Medical & Dental Sciences, Tokyo Medical & Dental University, Tokyo, Japan
| | - Michihisa Umetani
- Division of Pulmonary & Vascular Biology, Departments of Pediatrics & Pharmacology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| |
Collapse
|
9
|
McDonnell DP, Park S, Goulet MT, Jasper J, Wardell SE, Chang CY, Norris JD, Guyton JR, Nelson ER. Obesity, cholesterol metabolism, and breast cancer pathogenesis. Cancer Res 2014; 74:4976-82. [PMID: 25060521 PMCID: PMC4167494 DOI: 10.1158/0008-5472.can-14-1756] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Obesity and altered lipid metabolism are risk factors for breast cancer in pre- and post-menopausal women. These pathologic relationships have been attributed in part to the impact of cholesterol on the biophysical properties of cell membranes and to the influence of these changes on signaling events initiated at the membrane. However, more recent studies have indicated that the oxysterol 27-hydroxycholesterol (27HC), and not cholesterol per se, may be the primary biochemical link between lipid metabolism and cancer. The enzyme responsible for production of 27HC from cholesterol, CYP27A1, is expressed primarily in the liver and in macrophages. In addition, significantly elevated expression of this enzyme within breast tumors has also been observed. It is believed that 27HC, acting through the liver X receptor in macrophages and possibly other cells, is involved in maintaining organismal cholesterol homeostasis. It has also been shown recently that 27HC is an estrogen receptor agonist in breast cancer cells and that it stimulates the growth and metastasis of tumors in several models of breast cancer. These findings provide the rationale for the clinical evaluation of pharmaceutical approaches that interfere with cholesterol/27HC synthesis as a means to mitigate the impact of cholesterol on breast cancer pathogenesis. Cancer Res; 74(18); 4976-82. ©2014 AACR.
Collapse
Affiliation(s)
- Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina.
| | - Sunghee Park
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Matthew T Goulet
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Jeff Jasper
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Suzanne E Wardell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - John D Norris
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - John R Guyton
- Division of Endocrinology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Illinois
| |
Collapse
|
10
|
Nelson ER, Wardell SE, Jasper JS, Park S, Suchindran S, Howe MK, Carver NJ, Pillai RV, Sullivan PM, Sondhi V, Umetani M, Geradts J, McDonnell DP. 27-Hydroxycholesterol links hypercholesterolemia and breast cancer pathophysiology. Science 2013; 342:1094-8. [PMID: 24288332 DOI: 10.1126/science.1241908] [Citation(s) in RCA: 598] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Hypercholesterolemia is a risk factor for estrogen receptor (ER)-positive breast cancers and is associated with a decreased response of tumors to endocrine therapies. Here, we show that 27-hydroxycholesterol (27HC), a primary metabolite of cholesterol and an ER and liver X receptor (LXR) ligand, increases ER-dependent growth and LXR-dependent metastasis in mouse models of breast cancer. The effects of cholesterol on tumor pathology required its conversion to 27HC by the cytochrome P450 oxidase CYP27A1 and were attenuated by treatment with CYP27A1 inhibitors. In human breast cancer specimens, CYP27A1 expression levels correlated with tumor grade. In high-grade tumors, both tumor cells and tumor-associated macrophages exhibited high expression levels of the enzyme. Thus, lowering circulating cholesterol levels or interfering with its conversion to 27HC may be a useful strategy to prevent and/or treat breast cancer.
Collapse
Affiliation(s)
- Erik R Nelson
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
27-Hydroxycholesterol promotes cell-autonomous, ER-positive breast cancer growth. Cell Rep 2013; 5:637-45. [PMID: 24210818 DOI: 10.1016/j.celrep.2013.10.006] [Citation(s) in RCA: 275] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/11/2013] [Accepted: 10/02/2013] [Indexed: 01/10/2023] Open
Abstract
To date, estrogen is the only known endogenous estrogen receptor (ER) ligand that promotes ER+ breast tumor growth. We report that the cholesterol metabolite 27-hydroxycholesterol (27HC) stimulates MCF-7 cell xenograft growth in mice. More importantly, in ER+ breast cancer patients, 27HC content in normal breast tissue is increased compared to that in cancer-free controls, and tumor 27HC content is further elevated. Increased tumor 27HC is correlated with diminished expression of CYP7B1, the 27HC metabolizing enzyme, and reduced expression of CYP7B1 in tumors is associated with poorer patient survival. Moreover, 27HC is produced by MCF-7 cells, and it stimulates cell-autonomous, ER-dependent, and GDNF-RET-dependent cell proliferation. Thus, 27HC is a locally modulated, nonaromatized ER ligand that promotes ER+ breast tumor growth.
Collapse
|
12
|
Nelson ER, Wardell SE, McDonnell DP. The molecular mechanisms underlying the pharmacological actions of estrogens, SERMs and oxysterols: implications for the treatment and prevention of osteoporosis. Bone 2013; 53:42-50. [PMID: 23168292 PMCID: PMC3552054 DOI: 10.1016/j.bone.2012.11.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 10/19/2012] [Accepted: 11/12/2012] [Indexed: 12/12/2022]
Abstract
Estrogen therapy and hormone therapy are effective options for the prevention and treatment of osteoporosis, although because of their significant side effect profile, long term use for these applications is not recommended. Whereas SERMs (Selective Estrogen Receptor Modulators) exhibit a more favorable side effect profile, the currently available medicines in this class are substantially less effective in bone than classical estrogens. However, the results of substantial efforts that have gone into defining the mechanisms that underlie the pharmacology of estrogens, antiestrogens and SERMs have informed the development of the next generation of SERMs and have led to the development of TSECs (Tissue Selective Estrogen Complexes), a new class of ER-modulator. Further, the recent determination that the oxysterol 27-hydroxycholesterol functions as an endogenous SERM has highlighted an unexpected link between hypercholesterolemia and bone biology and must be considered in any discussions of ER-pharmacology. This review considers the most recent progress in our understanding of ER pharmacology and how this has and will be translated into new medicines for the treatment and prevention of osteoporosis.
Collapse
Affiliation(s)
- Erik R Nelson
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | | | | |
Collapse
|
13
|
Dong C, Zhao G, Zhong M, Yue Y, Wu L, Xiong S. RNA sequencing and transcriptomal analysis of human monocyte to macrophage differentiation. Gene 2013; 519:279-87. [PMID: 23458880 DOI: 10.1016/j.gene.2013.02.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Revised: 01/02/2013] [Accepted: 02/07/2013] [Indexed: 12/24/2022]
Abstract
Monocytes can be differentiated into macrophages in vivo and these cells play an important role in innate and adaptive immune responses. To reveal the global gene transcription change that occurs during monocyte to macrophage differentiation, we performed genome-wide RNA sequencing and analyses in human primary monocytes and monocyte-derived macrophages. We show that 1208 genes (with >twofold differences) were differentially expressed in macrophages compared with monocytes, including 800 upregulated and 408 downregulated genes. Gene ontology, pathway, and protein-protein interaction analyses indicated that the upregulated genes were related to macrophage functions in phagocytosis, metabolic processes, and cell cycle. The majority of downregulated genes comprised genes involved in the inflammatory response and locomotion. Genes encoding transcription regulatory factors, such as FOXO1, RUNX3, NF-κB1, and C/EBP δ, were highly expressed in monocytes and appeared to function in significant transcriptional repression, resulting in slight metabolic activity. Our transcriptome comparison between human monocytes and monocyte-derived macrophages using RNA sequencing revealed novel molecules and pathways associated with the differentiation process. These molecules and pathways may represent candidate targets involved in the pathophysiology of these important immune cells.
Collapse
Affiliation(s)
- Chunsheng Dong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, PR China.
| | | | | | | | | | | |
Collapse
|
14
|
Bianchi E, Scarinci F, Ripandelli G, Feher J, Pacella E, Magliulo G, Gabrieli CB, Plateroti R, Plateroti P, Mignini F, Artico M. Retinal pigment epithelium, age-related macular degeneration and neurotrophic keratouveitis. Int J Mol Med 2012; 31:232-42. [PMID: 23128960 DOI: 10.3892/ijmm.2012.1164] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 09/25/2012] [Indexed: 11/05/2022] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of impaired vision and blindness in the aging population. The aims of our studies were to identify qualitative and quantitative alterations in mitochondria in human retinal pigment epithelium (RPE) from AMD patients and controls and to test the protective effects of pigment epithelium-derived factor (PEDF), a known neurotrophic and antiangiogenic substance, against neurotrophic keratouveitis. Histopathological alterations were studied by means of morphometry, light and electron microscopy. Unexpectedly, morphometric data showed that the RPE alterations noted in AMD may also develop in normal aging, 10-15 years later than appearing in AMD patients. Reduced tear secretion, corneal ulceration and leukocytic infiltration were found in capsaicin (CAP)-treated rats, but this effect was significantly attenuated by PEDF. These findings suggest that PEDF accelerated the recovery of tear secretion and also prevented neurotrophic keratouveitis and vitreoretinal inflammation. PEDF may have a clinical application in inflammatory and neovascular diseases of the eye.
Collapse
Affiliation(s)
- Enrica Bianchi
- Department of Sensory Organs, University of Rome, La Sapienza, Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Yam KC, Okamoto S, Roberts JN, Eltis LD. Adventures inRhodococcus — from steroids to explosivesThis article is based on a presentation by Dr. Lindsay Eltis at the 60th Annual Meeting of the Canadian Society of Microbiologists in Hamilton, Ontario, 14 June 2010. Dr. Eltis was the recipient of the 2010 Norgen Biotek Corporation / CSM Award, an annual award sponsored by Norgen Biotek and the Canadian Society of Microbiologists intended to recognize outstanding scientific work in microbiology by a Canadian researcher. Can J Microbiol 2011; 57:155-68. [DOI: 10.1139/w10-115] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Rhodococcus is a genus of mycolic-acid-containing actinomycetes that utilize a remarkable variety of organic compounds as growth substrates. This degradation helps maintain the global carbon cycle and has increasing applications ranging from the biodegradation of pollutants to the biocatalytic production of drugs and hormones. We have been using Rhodococcus jostii RHA1 as a model organism to understand the catabolic versatility of Rhodococcus and related bacteria. Our approach is exemplified by the discovery of a cluster of genes specifying the catabolism of cholesterol. This degradation proceeds via β-oxidative degradation of the side chain and O2-dependent cleavage of steroid ring A in a process similar to bacterial degradation of aromatic compounds. The pathway is widespread in Actinobacteria and is critical to the pathogenesis of Mycobacterium tuberculosis , arguably the world’s most successful pathogen. The close similarity of some of these enzymes with biphenyl- and polychlorinated-biphenyl-degrading enzymes that we have characterized is facilitating inhibitor design. Our studies in RHA1 have also provided important insights into a number of novel metalloenzymes and their biosynthesis, such as acetonitrile hydratase (ANHase), a cobalt-containing enzyme with no significant sequence identity with characterized nitrile hydratases. Molecular genetic and biochemical studies have identified AnhE as a dimeric metallochaperone that delivers cobalt to ANHase, enabling its maturation in vivo. Other metalloenzymes we are characterizing include N-acetylmuramic acid hydroxylase, which catalyzes an unusual hydroxylation of the rhodococcal and mycobacterial peptidoglycan, and 2 RHA1 dye-decolorizing peroxidases. Using molecular genetic and biochemical approaches, we have demonstrated that one of these enzymes is involved in the degradation of lignin. Overall, our studies are providing fundamental insights into a range of catabolic processes that have a wide variety of applications.
Collapse
Affiliation(s)
- Katherine C. Yam
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Sachi Okamoto
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Joseph N. Roberts
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Lindsay D. Eltis
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
16
|
Driscoll MD, McLean KJ, Levy C, Mast N, Pikuleva IA, Lafite P, Rigby SEJ, Leys D, Munro AW. Structural and biochemical characterization of Mycobacterium tuberculosis CYP142: evidence for multiple cholesterol 27-hydroxylase activities in a human pathogen. J Biol Chem 2010; 285:38270-82. [PMID: 20889498 PMCID: PMC2992261 DOI: 10.1074/jbc.m110.164293] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 09/09/2010] [Indexed: 11/06/2022] Open
Abstract
The Mycobacterium tuberculosis cytochrome P450 enzyme CYP142 is encoded in a large gene cluster involved in metabolism of host cholesterol. CYP142 was expressed and purified as a soluble, low spin P450 hemoprotein. CYP142 binds tightly to cholesterol and its oxidized derivative cholest-4-en-3-one, with extensive shift of the heme iron to the high spin state. High affinity for azole antibiotics was demonstrated, highlighting their therapeutic potential. CYP142 catalyzes either 27-hydroxylation of cholesterol/cholest-4-en-3-one or generates 5-cholestenoic acid/cholest-4-en-3-one-27-oic acid from these substrates by successive sterol oxidations, with the catalytic outcome dependent on the redox partner system used. The CYP142 crystal structure was solved to 1.6 Å, revealing a similar active site organization to the cholesterol-metabolizing M. tuberculosis CYP125, but having a near-identical organization of distal pocket residues to the branched fatty acid oxidizing M. tuberculosis CYP124. The cholesterol oxidizing activity of CYP142 provides an explanation for previous findings that ΔCYP125 strains of Mycobacterium bovis and M. bovis BCG cannot grow on cholesterol, because these strains have a defective CYP142 gene. CYP142 is revealed as a cholesterol 27-oxidase with likely roles in host response modulation and cholesterol metabolism.
Collapse
Affiliation(s)
- Max D. Driscoll
- From the Manchester Interdisciplinary Biocentre, Faculty of Life Sciences, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Kirsty J. McLean
- From the Manchester Interdisciplinary Biocentre, Faculty of Life Sciences, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Colin Levy
- From the Manchester Interdisciplinary Biocentre, Faculty of Life Sciences, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Natalia Mast
- the Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio 444106, and
| | - Irina A. Pikuleva
- the Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio 444106, and
| | - Pierre Lafite
- the ICOA-UMR, CNRS 6005, Université d'Orléans, Rue de Chartres, 45067 Orléans, France
| | - Stephen E. J. Rigby
- From the Manchester Interdisciplinary Biocentre, Faculty of Life Sciences, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - David Leys
- From the Manchester Interdisciplinary Biocentre, Faculty of Life Sciences, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Andrew W. Munro
- From the Manchester Interdisciplinary Biocentre, Faculty of Life Sciences, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| |
Collapse
|
17
|
Capyk JK, Kalscheuer R, Stewart GR, Liu J, Kwon H, Zhao R, Okamoto S, Jacobs WR, Eltis LD, Mohn WW. Mycobacterial cytochrome p450 125 (cyp125) catalyzes the terminal hydroxylation of c27 steroids. J Biol Chem 2009; 284:35534-42. [PMID: 19846551 PMCID: PMC2790983 DOI: 10.1074/jbc.m109.072132] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 10/17/2009] [Indexed: 11/06/2022] Open
Abstract
Cyp125 (Rv3545c), a cytochrome P450, is encoded as part of the cholesterol degradation gene cluster conserved among members of the Mycobacterium tuberculosis complex. This enzyme has been implicated in mycobacterial pathogenesis, and a homologue initiates cholesterol catabolism in the soil actinomycete Rhodococcus jostii RHA1. In Mycobacterium bovis BCG, cyp125 was up-regulated 7.1-fold with growth on cholesterol. A cyp125 deletion mutant of BCG did not grow on cholesterol and accumulated 4-cholesten-3-one when incubated in the presence of cholesterol. Wild-type BCG grew on this metabolite. By contrast, a parallel cyp125 deletion mutation of M. tuberculosis H37Rv did not affect growth on cholesterol. Purified Cyp125 from M. tuberculosis, heterologously produced in R. jostii RHA1, bound cholesterol and 4-cholesten-3-one with apparent dissociation constants of 0.20 +/- 0.02 microM and 0.27 +/- 0.05 microm, respectively. When reconstituted with KshB, the cognate reductase of the ketosteroid 9alpha-hydroxylase, Cyp125 catalyzed the hydroxylation of these steroids. MS and NMR analyses revealed that hydroxylation occurred at carbon 26 of the steroid side chain, allowing unambiguous classification of Cyp125 as a steroid C26-hydroxylase. This study establishes the catalytic function of Cyp125 and, in identifying an important difference in the catabolic potential of M. bovis and M. tuberculosis, suggests that Cyp125 may have an additional function in pathogenesis.
Collapse
Affiliation(s)
- Jenna K. Capyk
- From the Departments of Biochemistry and Molecular Biology and
| | - Rainer Kalscheuer
- the Howard Hughes Medical Institute, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Gordon R. Stewart
- Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada and
| | - Jie Liu
- Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada and
| | - Hyukin Kwon
- Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada and
| | - Rafael Zhao
- From the Departments of Biochemistry and Molecular Biology and
| | - Sachi Okamoto
- Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada and
| | - William R. Jacobs
- the Howard Hughes Medical Institute, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Lindsay D. Eltis
- From the Departments of Biochemistry and Molecular Biology and
- Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada and
| | - William W. Mohn
- Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada and
| |
Collapse
|
18
|
Vantieghem K, Kissmeyer AM, De Haes P, Bouillon R, Segaert S. UVB-induced production of 1,25-dihydroxyvitamin D3 and vitamin D activity in human keratinocytes pretreated with a sterol delta7-reductase inhibitor. J Cell Biochem 2009; 98:81-92. [PMID: 16365879 DOI: 10.1002/jcb.20756] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The skin fulfills an important role in the vitamin D photo-endocrine system. Epidermis is not only the site of vitamin D3 photoproduction. In addition, epidermal keratinocytes contain the vitamin D receptor (VDR) and possess 25-hydroxylase and 1alpha-hydroxylase activity indicating that all components of the vitamin D system are present. We investigated whether these components cooperate in inducing vitamin D activity upon treatment with physiological UVB doses. Upon irradiation, 24-hydroxylase mRNA was induced in keratinocytes pretreated with a sterol Delta7-reductase inhibitor (BM15766) whereby the 7-dehydrocholesterol content increased by 300-fold. Transfection experiments with a vitamin D response element containing construct confirmed VDR-dependent gene activation. Furthermore, the UVB-dependent induction of 24-hydroxylase was blocked by the cytochrome-P450 inhibitor ketoconazole. The 24-hydroxylase inducing photoproduct was transferable to unirradiated keratinocytes by medium and cellular homogenates of UVB-irradiated, BM15766-pretreated cells and was identified as 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] by high-performance liquid chromatography with tandem mass spectrometric detection. Addition of vitamin D binding protein blunted UVB-induced 24-hydroxylase suggesting the possibility of a paracrine or autocrine role for 1,25(OH)2D3. In conclusion, epidermal keratinocytes can produce vitamin D3, convert it to 1,25(OH)2D3 and respond to it upon UVB irradiation in the absence of exogenous 7-dehydrocholesterol and therefore contain a unique and complete photo-endocrine vitamin D system.
Collapse
Affiliation(s)
- Katleen Vantieghem
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Gasthuisberg, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | | | | |
Collapse
|
19
|
27-Hydroxycholesterol: a potential endogenous regulator of estrogen receptor signaling. Trends Pharmacol Sci 2008; 29:510-4. [PMID: 18722677 DOI: 10.1016/j.tips.2008.07.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 07/14/2008] [Accepted: 07/22/2008] [Indexed: 11/21/2022]
Abstract
The selective estrogen receptor modulators (SERMs) are synthetic pharmaceuticals, the relative agonist and antagonist activities of which are not equivalent in all cells. Their discovery has raised the possibility that endogenous small molecules might exist that have similar properties and could have important physiological roles. In support of this hypothesis is the recent demonstration that the oxysterol 27-hydroxycholesterol (27HC) interacts with and modulates the transcriptional activity of both estrogen receptor (ER) subtypes and that the relative agonist and antagonist activity of 27HC is influenced by both cell and promoter context. Although there is limited information available on the role of 27HC in classical estrogen-responsive tissues, that which is available in animal models of cardiovascular disease and cellular models of breast cancer support a role for this ligand in ER signaling. These results provide an interesting potential link between cholesterol (and cholesterol metabolism) and ER function, the physiological and pathological importance of which remains to be determined.
Collapse
|
20
|
Liu H, Shi B, Huang CC, Eksarko P, Pope RM. Transcriptional diversity during monocyte to macrophage differentiation. Immunol Lett 2008; 117:70-80. [PMID: 18276018 DOI: 10.1016/j.imlet.2007.12.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Revised: 12/17/2007] [Accepted: 12/19/2007] [Indexed: 10/22/2022]
Abstract
Monocytes recruited into tissues from peripheral blood differentiate into macrophages, which are critical in the pathogenesis of many diseases. There is limited data concerning the global changes in the expression of genes during monocyte to macrophage differentiation, and how the patterns of change identify the mechanism contributing to macrophage differentiation or function. Employing microarray technology, we examined the transcriptional profile of in vitro adherence-induced differentiation of primary human monocytes into macrophages. We found the significant up regulation of genes contributing to the functions of macrophages, including those regulating to immunity and defense; lipid, fatty acid and steroid metabolism; cell adhesion, carbohydrate metabolism; amino acid metabolism and endocytosis. In contrast, the vast majority of transcription factors affected were down regulated during monocyte to macrophage differentiation, suggesting that transcriptional repression may be important for the transition from monocytes to macrophages. However, a limited number of transcription factors were up regulated, among these was C/EBPalpha, which may contribute to differentiation by regulating down stream genes, which are a characteristic of differentiated macrophages. These observations suggest that examination of the transcriptional profile in monocytes and macrophages in patients may identify relevant therapeutic targets in diseases mediated by macrophages.
Collapse
Affiliation(s)
- Hongtao Liu
- Northwestern University Feinberg School of Medicine, Division of Rheumatology, Chicago, IL 60611, United States
| | | | | | | | | |
Collapse
|
21
|
Vantieghem K, Overbergh L, Carmeliet G, De Haes P, Bouillon R, Segaert S. UVB-induced 1,25(OH)2D3 production and vitamin D activity in intestinal CaCo-2 cells and in THP-1 macrophages pretreated with a sterol Delta7-reductase inhibitor. J Cell Biochem 2006; 99:229-40. [PMID: 16598763 DOI: 10.1002/jcb.20910] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Epidermal keratinocytes are able to produce 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] and induce vitamin D activity upon UVB irradiation. To find out whether this property is keratinocyte specific, we investigated this characteristic in two other cell types, namely intestinal CaCo-2 cells and the macrophage-like differentiated THP-1 cells. THP-1 macrophages and preconfluent CaCo-2 cells contain the vitamin D receptor (VDR), possess 25-hydroxylase (CYP2R1 and CYP27A1) and 1alpha-hydroxylase (CYP27B1) activity, and survive the low UVB doses essential for vitamin D3 photoproduction. Upon irradiation, 24-hydroxylase (CYP24) mRNA is induced in both cell types pretreated with the sterol Delta7-reductase inhibitor BM15766 whereby the 7-dehydrocholesterol (7-DHC) content was increased. Transfection studies in CaCo-2 cells with a vitamin D response element-containing construct revealed the involvement of the VDR in this UVB-dependent CYP24 induction. The CYP24 inducing activity in BM15766-pretreated UVB-irradiated CaCo-2 cells and THP-1 macrophages was identified as 1,25(OH)2D3 by combined high-performance liquid chromatography radioimmunoassay. Addition of vitamin D binding protein to the CaCo-2 cells attenuated UVB-induced CYP24 induction suggesting the possibility of a paracrine or autocrine role for the photoproduced 1,25(OH)2D3. In conclusion, preconfluent CaCo-2 cells and THP-1 macrophages are able to induce vitamin D activity upon UVB irradiation and hence combine all parts of the vitamin D photoendocrine system, a characteristic which is therefore not keratinocyte specific.
Collapse
Affiliation(s)
- Katleen Vantieghem
- Laboratory for Experimental Medicine and Endocrinology (LEGENDO), Gasthuisberg, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
22
|
Vantieghem K, De Haes P, Bouillon R, Segaert S. Dermal fibroblasts pretreated with a sterol Δ7-reductase inhibitor produce 25-hydroxyvitamin D3 upon UVB irradiation. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2006; 85:72-8. [PMID: 16824767 DOI: 10.1016/j.jphotobiol.2006.05.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Revised: 05/11/2006] [Accepted: 05/11/2006] [Indexed: 11/29/2022]
Abstract
As dermis is a physiological site of vitamin D3 photoproduction, the photo-endocrine vitamin D3 system was studied in dermal fibroblasts. Dermal fibroblasts contain the vitamin D receptor and induce 1alpha,25-dihydroxyvitamin D3-24-hydroxylase [CYP24] mRNA upon stimulation with 1,25-dihydroxyvitamin D3 [1,25(OH)2D3]. In addition, dermal fibroblasts contain mRNA of the vitamin D3-25-hydroxylases (CYP2R1 and CYP27A1). However, we could not detect any 25-hydroxyvitamin D3 [25OHD3]-1alpha-hydroxylase mRNA in dermal fibroblasts and no CYP24 mRNA was induced upon ultraviolet [UVB] irradiation, even when endogenous 7-dehydrocholesterol content was elevated by pretreatment with the sterol Delta7-reductase inhibitor BM15766. Nevertheless, dermal fibroblasts produce inactive vitamin D3 metabolites that can be activated by epidermal keratinocytes as CYP24 mRNA is induced in epidermal keratinocytes but not in dermal fibroblasts after transfer of medium or cellular suspensions from BM15766-pretreated, UVB-irradiated fibroblasts. This CYP24 induction was UVB-dose dependent and was inhibited by ketoconazole. As revealed in a competitive binding assay, BM15766-pretreated dermal fibroblasts are able to produce 25OHD3 upon UVB irradiation, but no 1,25(OH)2D3 was detected via combined high-performance liquid chromatography radioimmunoassay. The physiological relevance of dermal vitamin D3 photoproduction and its subsequent conversion into 25OHD3 remains elusive.
Collapse
Affiliation(s)
- Katleen Vantieghem
- Laboratorium voor Experimentele Geneeskunde en Endocrinologie (LEGENDO), Gasthuisberg, Katholieke Universiteit Leuven, Herestraat 49, 3000 Leuven, Belgium
| | | | | | | |
Collapse
|
23
|
Gottfried E, Rehli M, Hahn J, Holler E, Andreesen R, Kreutz M. Monocyte-derived cells express CYP27A1 and convert vitamin D3 into its active metabolite. Biochem Biophys Res Commun 2006; 349:209-13. [PMID: 16930540 DOI: 10.1016/j.bbrc.2006.08.034] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Accepted: 08/07/2006] [Indexed: 11/22/2022]
Abstract
CYP27A1 catalyses hydroxylations in the biosynthesis of bile acids and the bioactivation of vitamin D3. We investigated the expression of CYP27A1 in human monocytes, monocyte-derived macrophages, and dendritic cells on mRNA and protein levels as well as its enzymatic activity in comparison with the expression of CYP27B1 and CYP24A1. Macrophages showed a strong expression of CYP27A1, whereas monocytes and dendritic cells expressed low levels of CYP27A1 mRNA. Immunohistochemistry revealed CYP27A1 and CYP27B1 protein expression in macrophages. Accordingly, macrophages converted vitamin D3 into the active metabolite 1,25(OH)2D3. Dendritic cells also metabolized vitamin D3 although to a lesser extent. This could be due to the high expression of CYP24A1, the enzyme that degrades 25(OH)D3 and 1,25(OH)2D3. Our results show that macrophages and dendritic cells are capable to perform both hydroxylation steps of the vitamin D3 metabolism suggesting a possible role of local 1,25(OH)2D3 synthesis by myeloid cells in the skin and gut.
Collapse
Affiliation(s)
- Eva Gottfried
- Department of Hematology and Oncology, University of Regensburg, Franz-Josef Strauss Allee 11, 93042 Regensburg, Germany
| | | | | | | | | | | |
Collapse
|
24
|
Llaverias G, Rebollo A, Pou J, Vázquez-Carrera M, Sánchez RM, Laguna JC, Alegret M. Effects of rosiglitazone and atorvastatin on the expression of genes that control cholesterol homeostasis in differentiating monocytes. Biochem Pharmacol 2006; 71:605-14. [PMID: 16386711 DOI: 10.1016/j.bcp.2005.11.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2005] [Revised: 11/23/2005] [Accepted: 11/23/2005] [Indexed: 10/25/2022]
Abstract
We studied the effects of 5 microM atorvastatin, 2 microM rosiglitazone and their combination on intracellular cholesterol levels and on the expression of genes controlling cholesterol trafficking in human monocytes during their differentiation into macrophages. Our results show that treatment with rosiglitazone caused an increase in CD36 mRNA and protein levels (2.7- and 2.9-fold, P<0.001), but significantly induced the expression of most genes related to cholesterol efflux: ABCA1 mRNA (23%, P<0.05) and protein (2.4-fold, P<0.05), apo E protein (2.4-fold, P<0.05), caveolin-1 mRNA (2.6-fold, P<0.001) and SR-BI mRNA (1.9-fold, P<0.001) and protein (3-fold, P<0.01). As a consequence, rosiglitazone treatment reduced intracellular free cholesterol levels by 22% (P<0.01). Treatment with 5 microM atorvastatin caused the opposite effect on the expression of cholesterol efflux-related genes, which was generally reduced: ABCA1 mRNA (71%, P<0.05), apo E mRNA (46%, P<0.001) and protein (5.6-fold, P<0.001), and CYP27 mRNA (15%, P<0.05). Despite these reductions, intracellular total and free cholesterol levels were also reduced by 30% (P<0.01), an effect that can be attributed to the inhibition of de novo cholesterol synthesis by the statins. The combination of rosiglitazone with atorvastatin attenuated CD36 induction, and caused reductions similar to those caused by the statin alone on the expression of genes involved in cholesterol efflux and on intracellular cholesterol levels.
Collapse
Affiliation(s)
- Gemma Llaverias
- Unitat de Farmacologia, Departament de Farmacologia i Química Terapèutica, Facultat de Farmàcia, Universitat de Barcelona, Av. Diagonal 643. 08028 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
25
|
Quinn C, Jessup W, Wong J, Kritharides L, Brown A. Expression and regulation of sterol 27-hydroxylase (CYP27A1) in human macrophages: a role for RXR and PPARgamma ligands. Biochem J 2005; 385:823-30. [PMID: 15533057 PMCID: PMC1134759 DOI: 10.1042/bj20041776] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CYP27A1 (sterol 27-hydroxylase) catalyses an important sterol elimination pathway in the human macrophage, and consequently may protect against atherosclerosis. We studied the expression and regulation of CYP27A1 in a human macrophage-like cell-line, THP-1, and primary HMDMs (human monocyte-derived macrophages). In both macrophage cell types, we found that CYP27A1 expression is independent of cellular cholesterol levels and of LXR (liver X receptor)-dependent control of transcription. However, the RXR (retinoid X receptor) ligand, 9-cis-retinoic acid, upregulates CYP27A1 expression. Of the RXR heterodimeric partners tested, PPAR (peroxisome-proliferator-activated receptor) gamma ligands significantly increased CYP27A1 mRNA levels. Its reversal by a PPARgamma antagonist demonstrated the specificity of this effect. Interestingly, HMDMs express markedly higher levels of CYP27A1 than THP-1 macrophages, and this difference was reflected in both protein levels and enzyme activities between the two cell types. In conclusion, stimulation of CYP27A1 by PPARgamma may represent a key previously unrecognized mechanism by which PPARgamma protects against atherosclerosis.
Collapse
Affiliation(s)
- Carmel M. Quinn
- *Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- †Centre for Vascular Research, Department of Haematology, Prince of Wales Hospital, Barker Street, Randwick, NSW 2031, Australia
| | - Wendy Jessup
- *Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- †Centre for Vascular Research, Department of Haematology, Prince of Wales Hospital, Barker Street, Randwick, NSW 2031, Australia
- To whom correspondence should be addressed at the Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia (email )
| | - Jenny Wong
- ‡School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Leonard Kritharides
- *Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- †Centre for Vascular Research, Department of Haematology, Prince of Wales Hospital, Barker Street, Randwick, NSW 2031, Australia
- §Department of Cardiology, Concord Hospital, Hospital Road, Concord, NSW 2139, Australia
| | - Andrew J. Brown
- ‡School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
26
|
Feher J, Kovacs I, Artico M, Cavallotti C, Papale A, Balacco Gabrieli C. Mitochondrial alterations of retinal pigment epithelium in age-related macular degeneration. Neurobiol Aging 2005; 27:983-93. [PMID: 15979212 DOI: 10.1016/j.neurobiolaging.2005.05.012] [Citation(s) in RCA: 270] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2004] [Revised: 05/08/2005] [Accepted: 05/19/2005] [Indexed: 12/31/2022]
Abstract
Mitochondrial dysfunctions have been implicated in the pathophysiology of several age-related diseases including age-related macular degeneration (AMD), a progressive neurodegenerative disease affecting primarily the retinal pigment epithelium (RPE). The aims of our electron microscopic and morphometric studies were to reveal qualitative and quantitative alterations of mitochondria in human RPE from AMD and from age- and sex-matched controls. With increasing age a significant decrease in number and area of mitochondria, as well as loss of cristae and matrix density were found in both AMD and control specimens. These decreases were significantly greater in AMD than in normal aging. Alterations of mitochondria were accompanied by proliferation of peroxisomes and lipofuscin granules in both AMD and control specimens, although the difference between groups was significant only for peroxisomes. Unexpectedly, morphometric data showed that the RPE alterations seen in AMD may also develop in normal aging, 10-15 years after appearing in AMD patients. These findings suggest that (i) the severity of mitochondrial and peroxisomal alterations are different between AMD and normal aging, and (ii) the timing of damage to RPE may be critical for the development of AMD. We conclude that besides the well-documented age-related changes in mitochondrial DNA, alterations of mitochondrial membranes may also play a role in the pathogenesis of AMD. These membranes could be a new target for treatment of AMD and other age-related diseases.
Collapse
Affiliation(s)
- Janos Feher
- Opthalmic Neuroscience Program, Department of Ophthalmology, University of Rome La Sapienza, Via Lombardia, 23/c, 00187 Rome, Italy.
| | | | | | | | | | | |
Collapse
|
27
|
Hansson M, Wikvall K, Babiker A. Regulation of sterol 27-hydroxylase in human monocyte-derived macrophages: up-regulation by transforming growth factor β1. Biochim Biophys Acta Mol Cell Biol Lipids 2005; 1687:44-51. [PMID: 15708352 DOI: 10.1016/j.bbalip.2004.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2004] [Revised: 10/04/2004] [Accepted: 11/03/2004] [Indexed: 01/22/2023]
Abstract
Regulatory mechanisms for human CYP27A1 enzyme have not yet been fully investigated. Our approach was to add different hormones and cytokines to cultured human monocyte-derived macrophages, and assess the effects on the CYP27A1 by measuring the production of 27-hydroxylated cholesterol in the media. Of the different hormones and cytokines tested, only transforming growth factor beta1 (TGF-beta1) had a clear effect on CYP27A1. Further experiments showed a significant increase in 27-hydroxylated cholesterol products (27-hydroxycholesterol and 3beta-hydroxy-5-cholestenoic acid). A concomitant increase in CYP27A1 mRNA levels was also seen and this positive effect was confirmed using a human CYP27A1 luciferase reporter gene expressed in HepG2 cells. Experiments with progressive deletion/luciferase reporter gene constructs indicated that a TGF-beta1 responsive sequence might be localized in a region about 400 bp upstream of the CYP27A1 translation start. The possibility is discussed that induction of CYP27A1 by TGF-beta1 may be responsible for some of the anti-atherogenic properties of this cytokine.
Collapse
Affiliation(s)
- Magnus Hansson
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska University Hospital at Huddinge, SE-141 86 Stockholm, Sweden
| | | | | |
Collapse
|
28
|
Szanto A, Benko S, Szatmari I, Balint BL, Furtos I, Rühl R, Molnar S, Csiba L, Garuti R, Calandra S, Larsson H, Diczfalusy U, Nagy L. Transcriptional regulation of human CYP27 integrates retinoid, peroxisome proliferator-activated receptor, and liver X receptor signaling in macrophages. Mol Cell Biol 2004; 24:8154-66. [PMID: 15340076 PMCID: PMC515045 DOI: 10.1128/mcb.24.18.8154-8166.2004] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cholesterol uptake and efflux are key metabolic processes associated with macrophage physiology and atherosclerosis. Peroxisome proliferator-activated receptor gamma (PPARgamma) and liver X receptor alpha (LXRalpha) have been linked to the regulation of these processes. It remains to be identified how activation of these receptors is connected and regulated by endogenous lipid molecules. We identified CYP27, a p450 enzyme, as a link between retinoid, PPARgamma, and LXR signaling. We show that the human CYP27 gene is under coupled regulation by retinoids and ligands of PPARs via a PPAR-retinoic acid receptor response element in its promoter. Induction of the enzyme's expression results in an increased level of 27-hydroxycholesterol and upregulation of LXR-mediated processes. Upregulated CYP27 activity also leads to LXR-independent elimination of CYP27 metabolites as an alternative means of cholesterol efflux. Moreover, human macrophage-rich atherosclerotic lesions have an increased level of retinoid-, PPARgamma-, and LXR-regulated gene expression and also enhanced CYP27 levels. Our findings suggest that nuclear receptor-regulated CYP27 expression is likely to be a key integrator of retinoic acid receptor-PPARgamma-LXR signaling, relying on natural ligands and contributing to lipid metabolism in macrophages.
Collapse
Affiliation(s)
- Attila Szanto
- Department of Biochemistry and Molecular Biology, Research Center for Molecular Medicine, University of Debrecen, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Meaney S, Bonfield TL, Hansson M, Babiker A, Kavuru MS, Thomassen MJ. Serum cholestenoic acid as a potential marker of pulmonary cholesterol homeostasis: increased levels in patients with pulmonary alveolar proteinosis. J Lipid Res 2004; 45:2354-60. [PMID: 15466366 DOI: 10.1194/jlr.m400302-jlr200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The conversion of cholesterol into the more polar metabolites 27-hydroxycholesterol (27-OH) and cholestenoic acid by the cytochrome P450 sterol 27-hydroxylase is a cholesterol-removal mechanism used by almost all cells. Most of the cholestenoic acid present in the circulation originates from the lung, and it has been suggested that sterol 27-hydroxylase is of particular importance for cholesterol homeostasis in this organ. As an example of pulmonary cholesterol accumulation, a known disorder of surfactant homeostasis, pulmonary alveolar proteinosis (PAP), was studied. Analysis of bronchoalveolar lavage fluid from PAP patients revealed a significant accumulation of the cholesterol metabolites cholestenoic acid and 27-OH. This pattern was recapitulated in serum, with a significant increase in the levels of both cholestenoic acid (P=0.003) and 27-OH (P=0.017) in PAP patients compared with healthy controls. Analysis of PAP alveolar macrophages did not reveal a significant change in mRNA expression levels of either sterol 27-hydroxylase or the cholesterol-esterifying enzyme acyl-CoA:cholesterol acyltransferase-1. These results are consistent with the contention that substrate availability, rather than enzyme expression, is the key factor in regulating the production of cholestenoic acid by the lung and that serum cholestenoic acid may be a marker of pulmonary cholesterol homeostasis.
Collapse
Affiliation(s)
- Steve Meaney
- Division of Clinical Chemistry, Karolinska Institutet, Huddinge University Hospital, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|