1
|
Hoffmann JP, Liu JA, Seddu K, Klein SL. Sex hormone signaling and regulation of immune function. Immunity 2023; 56:2472-2491. [PMID: 37967530 DOI: 10.1016/j.immuni.2023.10.008] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/25/2023] [Accepted: 10/14/2023] [Indexed: 11/17/2023]
Abstract
Immune responses to antigens, including innocuous, self, tumor, microbial, and vaccine antigens, differ between males and females. The quest to uncover the mechanisms for biological sex differences in the immune system has intensified, with considerable literature pointing toward sex hormonal influences on immune cell function. Sex steroids, including estrogens, androgens, and progestins, have profound effects on immune function. As such, drastic changes in sex steroid concentrations that occur with aging (e.g., after puberty or during the menopause transition) or pregnancy impact immune responses and the pathogenesis of immune-related diseases. The effect of sex steroids on immunity involves both the concentration of the ligand and the density and distribution of genomic and nongenomic receptors that serve as transcriptional regulators of immune cellular responses to affect autoimmunity, allergy, infectious diseases, cancers, and responses to vaccines. The next frontier will be harnessing these effects of sex steroids to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Joseph P Hoffmann
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jennifer A Liu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kumba Seddu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
2
|
Ortega RA, Bressman SB, Raymond D, Ozelius LJ, Katsnelson V, Leaver K, Swan MC, Shanker V, Miravite J, Wang C, Bennett SAL, Saunders-Pullman R. Differences in Sex-Specific Frequency of Glucocerebrosidase Variant Carriers and Familial Parkinsonism. Mov Disord 2022; 37:2217-2225. [PMID: 36054306 PMCID: PMC9669136 DOI: 10.1002/mds.29197] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/29/2022] [Accepted: 07/21/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Although men and women with the LRRK2 G2019S variant appear to be equally likely to have Parkinson's disease (PD), the sex-distribution among glucocerebrosidase (GBA) variant carriers with PD, including limited to specific variant severities of GBA, is not well understood. Further, the sex-specific genetic contribution to PD without a known genetic variant is controversial. OBJECTIVES To better understand sex differences in genetic contribution to PD, especially sex-specific frequencies among GBA variant carriers with PD (GBA PD) and LRRK2-G2019S variant carriers with PD (LRRK2 PD). METHODS We assess differences in the sex-specific frequency in GBA PD, including in subsets of GBA variant severity, LRRK2 PD, and idiopathic PD in an Ashkenazi Jewish cohort with PD. Further, we expand prior work evaluating differences in family history of parkinsonism. RESULTS Both idiopathic PD (267/420 men, 63.6%) (P < 0.001) and GBA PD overall (64/107, 59.8%) (P = 0.042) were more likely to be men, whereas no difference was seen in LRRK2 PD (50/99, 50.5%) and LRRK2/GBA PD (5/10, 50%). However, among GBA PD probands, severe variant carriers were more likely to be women (15/19 women, 79.0%) (P = 0.005), whereas mild variant carriers (44/70 men, 62.9%) (P = 0.039) and risk-variant carriers (15/17 men, 88.2%) (P = 0.001) were more likely to be men. CONCLUSIONS Our study demonstrates that the male-sex predominance present in GBA PD overall was not consistent across GBA variant severities, and a female-sex predominance was present among severe GBA variant carriers. Therefore, research and trial designs for PD should consider sex-specific differences, including across GBA variant severities. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Roberto A Ortega
- Department of Neurology, Mount Sinai Beth Israel, and Icahn School of Medicine, New York, New York, USA
| | - Susan B Bressman
- Department of Neurology, Mount Sinai Beth Israel, and Icahn School of Medicine, New York, New York, USA
| | - Deborah Raymond
- Department of Neurology, Mount Sinai Beth Israel, and Icahn School of Medicine, New York, New York, USA
| | - Laurie J Ozelius
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Viktoriya Katsnelson
- Department of Neurology, Mount Sinai Beth Israel, and Icahn School of Medicine, New York, New York, USA
| | - Katherine Leaver
- Department of Neurology, Mount Sinai Beth Israel, and Icahn School of Medicine, New York, New York, USA
| | - Matthew C Swan
- Department of Neurology, Mount Sinai Beth Israel, and Icahn School of Medicine, New York, New York, USA
| | - Vicki Shanker
- Department of Neurology, Mount Sinai Beth Israel, and Icahn School of Medicine, New York, New York, USA
| | - Joan Miravite
- Department of Neurology, Mount Sinai Beth Israel, and Icahn School of Medicine, New York, New York, USA
| | - Cuiling Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Steffany A L Bennett
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Ottawa Institute of Systems Biology, University of Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Rachel Saunders-Pullman
- Department of Neurology, Mount Sinai Beth Israel, and Icahn School of Medicine, New York, New York, USA
| |
Collapse
|
3
|
Albogami S, Hassan A, Abdel-Aziem SH, Alotaibi S, Althobaiti F, El-Shehawi A, Alnefaie A, Alhamed RA. Effects of combination of obesity, diabetes, and hypoxia on inflammatory regulating genes and cytokines in rat pancreatic tissues and serum. PeerJ 2022; 10:e13990. [PMID: 36213511 PMCID: PMC9541627 DOI: 10.7717/peerj.13990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/11/2022] [Indexed: 01/19/2023] Open
Abstract
Background Obesity and diabetes are becoming increasingly prevalent around the world. Inflammation, oxidative stress, insulin resistance, and glucose intolerance are linked to both obesity and type 2 diabetes, and these disorders are becoming major public health issues globally. Methods This study evaluated the effects of obesity, diabetes, and hypoxia on the levels of pro- and anti-inflammatory cytokines in rats. We divided 120 Wistar rats in two groups, male and female, each including six subgroups: control (CTRL), obese (high-fat diet (HFD)), diabetic (streptozotocin (STZ)-treated), hypoxic (HYX), obese + diabetic (HFD/STZ), and obese + diabetic + hypoxic (HFD/STZ/HYX). We examined the levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-6, IL10, and leptin in pancreatic tissues and serum. Results No significant difference was observed in serum levels of cholesterol, triglycerides, and low-density lipoprotein (LDL) between HYX and CTRL in either sex. However, they were significantly increased, whereas high-density lipoprotein (HDL) was significantly decreased in HFD, STZ, HFD/STZ, and HFD/STZ/HPX compared with CTRL in both sexes. The expression of Tnf-α, Il6, and Lep was significantly upregulated in all subgroups compared with CTRL in both sexes. STZ and HYX showed no significant differences in the expression of these genes between sexes, whereas Tnf-α and Il6 were upregulated in male HFD, HFD/STZ, and HFD/STZ/HYX compared with females. Protein levels showed similar patterns. Combination subgroups, either in the absence or presence of hypoxia, frequently exhibited severe necrosis of endocrine components in pancreatic lobules. The combination of obesity, diabetes, and hypoxia was associated with inflammation, which was verified at the histopathological level.
Collapse
Affiliation(s)
- Sarah Albogami
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia,High Altitude Research Center, Taif University, Taif, Saudi Arabia
| | - Aziza Hassan
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia,High Altitude Research Center, Taif University, Taif, Saudi Arabia
| | - Sekena H. Abdel-Aziem
- Cell Biology Department, Genetic Engineering and Biotechnology Division, National Research Centre, Cairo, Egypt
| | - Saqer Alotaibi
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia,High Altitude Research Center, Taif University, Taif, Saudi Arabia
| | - Fayez Althobaiti
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia,High Altitude Research Center, Taif University, Taif, Saudi Arabia
| | - Ahmed El-Shehawi
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia,High Altitude Research Center, Taif University, Taif, Saudi Arabia
| | - Alaa Alnefaie
- High Altitude Research Center, Taif University, Taif, Saudi Arabia,Department of Medical Services, King Faisal Medical Complex, Taif, Saudi Arabia
| | - Reem Abdulla Alhamed
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia,High Altitude Research Center, Taif University, Taif, Saudi Arabia
| |
Collapse
|
4
|
Kiefer ZE, Studer JM, Chipman AL, Adur MK, Mainquist-Whigham C, Gabler NK, Keating AF, Ross JW. Circulating biomarkers associated with pelvic organ prolapse risk in late gestation sows. J Anim Sci 2021; 99:6316206. [PMID: 34228800 PMCID: PMC8378218 DOI: 10.1093/jas/skab207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/05/2021] [Indexed: 12/22/2022] Open
Abstract
Sow mortality, as the result of pelvic organ prolapse (POP), has been increasing in the last decade in the U.S. swine industry. The objective of this study was to identify potential biological markers associated with risk of POP in sows. We hypothesized that sows differing in perineal score (PS) from PS1–PS3 (PS1—a presumed low POP risk; PS2—a presumed moderate POP risk; and PS3—a presumed high POP risk) would differ in circulatory biomarkers of inflammation and hormonal profiles. On gestation week 15, 2,864 individual sows were assigned a PS, and subsequently, 1.0%, 2.7%, and 23.4% of PS1, PS2, or PS3 sows, respectively, experienced POP. During PS assignment at days 107–116 of gestation, blood samples were collected from sows on two farms of similar genetics, feed sources, and health status. Whole blood was subjected to complete blood count (CBC) analysis (n = 212) and steroid hormones were measured in serum from a subset (n = 110) of animals assigned PS3 parity matched to PS1. Lipopolysaccharide-binding protein (LBP), tumor necrosis factor-alpha (TNF-α), haptoglobin, C-reactive protein (CRP), and creatine kinase (CK) levels were also evaluated. Complete blood count analysis revealed decreased (P ≤ 0.05) mean platelet volume (3.9%), lymphocytes (6.5%), and monocytes (7.5%) in PS3 compared to PS1 sows. Increased (P ≤ 0.02) abundance of androstenedione (13.4%), androsterone (18.2%), estrone (24.8%), and 17β-estradiol (26.2%) was observed in PS3 compared to PS1 sows. Additionally, a 25.8% increase (P = 0.04) in LBP in PS3 compared to PS1 sows was observed. Many dynamic physiological changes occur in sows during late gestation as they approach farrowing. The data presented herein demonstrate that distinct differences in concentrations of circulating biomarkers exist between late gestation sows at high or low risk for POP and may serve as a useful tool for understanding the etiology of POP and evaluation of mitigation strategies.
Collapse
Affiliation(s)
- Zoë E Kiefer
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Jamie M Studer
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | | | - Malavika K Adur
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | | | - Nicholas K Gabler
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Aileen F Keating
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Jason W Ross
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA.,Iowa Pork Industry Center, Ames, IA 50011, USA
| |
Collapse
|
5
|
Rastgar S, Alijani Ardeshir R, Zabihi E, Movahedinia A, Salati AP. Immunotoxicity of estrogen and nonylphenol on apoptosis and expression of ERs in goldfish macrophage: Opening new avenue for discovering the role of experimental model systems and sexes. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 209:159-167. [PMID: 30780113 DOI: 10.1016/j.aquatox.2019.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/08/2019] [Indexed: 06/09/2023]
Abstract
The expression of estrogen receptors (ERs) and their roles in important cell processes such as apoptosis in the macrophages exposed to estrogen/xenoestrogen have remained a complex secret. This study focused on the expression of estrogen receptors (ERs) and the stimulation of apoptosis in the macrophages from the two sexes of goldfish (Carassius auratus) exposed to 17-βestradiol (E2) and nonylphenol (NP) under in vivo and in vitro conditions. For the in vivo experiment, fish were exposed to NP (10-6 M and 10-7 M) and E2 (10-6 M) for 24 days. Then, the head kidney macrophages from the male and the female goldfish were isolated and assayed. For the in vitro experiments, the macrophages derived from the two sexes were cultured in L-15 medium and exposed to E2 (150 nM) and NP (10 nM and 150 nM) for 3 days. The results showed that the three isoforms of ERs (ERα, ERβ1, ERβ2) were expressed in the goldfish macrophages. After the exposure of macrophages to NP and E2, sex-specific increase of ERs expression and apoptosis were observed (P < 0.05). The expression of ERα after NP treatment showed the highest alteration, with the response being concentration-dependent. The most alteration of ERs expression were observed in the in vivo experiment. This study provides insight to understand how exposure of the goldfish macrophages to E2 and NP can up-regulate the transcript levels of estrogen receptor subtypes and stimulate apoptosis.
Collapse
Affiliation(s)
- Sara Rastgar
- Department of Marine Biology, Faculty of Marine Sciences, Khorramshahr University of Marine Science and Technology, P.O. Box 669, Khorramshahr, Iran
| | - Rashid Alijani Ardeshir
- Department of Marine Biology, Faculty of Marine Sciences, Khorramshahr University of Marine Science and Technology, P.O. Box 669, Khorramshahr, Iran
| | - Ebrahim Zabihi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abdolali Movahedinia
- Department of Marine Biology, Faculty of Marine Sciences, University of Mazandaran, Babolsar, Iran
| | - Amir Parviz Salati
- Department of Fisheries, Faculty of Marine Natural resources, Khorramshahr University of Marine Science and Technology, P.O. Box 669, Khorramshahr, Iran.
| |
Collapse
|
6
|
Mancera KF, Besson M, Lisle A, Allavena R, Phillips CJ. The effects of mining machinery noise of different amplitudes on the behaviour, faecal corticosterone and tissue morphology of wild mice ( Mus musculus ). Appl Anim Behav Sci 2018. [DOI: 10.1016/j.applanim.2018.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
7
|
Fan W, Xu Y, Liu Y, Zhang Z, Lu L, Ding Z. Obesity or Overweight, a Chronic Inflammatory Status in Male Reproductive System, Leads to Mice and Human Subfertility. Front Physiol 2018; 8:1117. [PMID: 29354072 PMCID: PMC5758580 DOI: 10.3389/fphys.2017.01117] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/18/2017] [Indexed: 12/18/2022] Open
Abstract
Obesity is frequently accompanied with chronic inflammation over the whole body and is always associated with symptoms that include those arising from metabolic and vascular alterations. On the other hand, the chronic inflammatory status in the male genital tract may directly impair spermatogenesis and is even associated with male subfertility. However, it is still unclear if the chronic inflammation induced by obesity damages spermatogenesis in the male genital tract. To address this question, we used a high fat diet (HFD) induced obese mouse model and recruited obese patients from the clinic. We detected increased levels of tumor necrosis factor (TNF-α), interleukin-6 (IL-6), and NOD-like receptor family pyrin domain containing-3 (NLRP3) in genital tract tissues including testis, epididymis, seminal vesicle, prostate, and serum from obese mice. Meanwhile, the levels of immunoglobulin G (IgG) and corticosterone were significantly higher than those in the control group in serum. Moreover, signal factors regulated by TNF-α, i.e., p38, nuclear factor-κB (NF-κB), Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK), and their phosphorylated status, and inflammasome protein NLRP3 were expressed at higher levels in the testis. For overweight and obese male patients, the increased levels of TNF-α and IL-6 were also observed in their seminal plasma. Furthermore, there was a positive correlation between the TNF-α and IL-6 levels and BMI whereas they were inversely correlated with the sperm concentration and motility. In conclusion, impairment of male fertility may stem from a chronic inflammatory status in the male genital tract of obese individuals.
Collapse
Affiliation(s)
- Weimin Fan
- Shanghai Key Laboratory for Reproductive Medicine, Department of Histology Embryology, Genetics and Developmental Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Reproductive Medicine Center, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yali Xu
- Shanghai Key Laboratory for Reproductive Medicine, Department of Histology Embryology, Genetics and Developmental Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Liu
- Shanghai Key Laboratory for Reproductive Medicine, Department of Histology Embryology, Genetics and Developmental Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhengqing Zhang
- The Laboratory of Clinical Medicine, Shanghai No.9 People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liming Lu
- Laboratory of Immune Regulation, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhide Ding
- Shanghai Key Laboratory for Reproductive Medicine, Department of Histology Embryology, Genetics and Developmental Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
8
|
Al-Tarrah K, Moiemen N, Lord JM. The influence of sex steroid hormones on the response to trauma and burn injury. BURNS & TRAUMA 2017; 5:29. [PMID: 28920065 PMCID: PMC5597997 DOI: 10.1186/s41038-017-0093-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 07/19/2017] [Indexed: 12/24/2022]
Abstract
Trauma and related sequelae result in disturbance of homeostatic mechanisms frequently leading to cellular dysfunction and ultimately organ and system failure. Regardless of the type and severity of injury, gender dimorphism in outcomes following trauma have been reported, with females having lower mortality than males, suggesting that sex steroid hormones (SSH) play an important role in the response of body systems to trauma. In addition, several clinical and experimental studies have demonstrated the effects of SSH on the clinical course and outcomes following injury. Animal studies have reported the ability of SSH to modulate immune, inflammatory, metabolic and organ responses following traumatic injury. This indicates that homeostatic mechanisms, via direct and indirect pathways, can be maintained by SSH at local and systemic levels and hence result in more favourable prognosis. Here, we discuss the role and mechanisms by which SSH modulates the response of the body to injury by maintaining various processes and organ functions. Such properties of sex hormones represent potential novel therapeutic strategies and further our understanding of current therapies used following injury such as oxandrolone in burn-injured patients.
Collapse
Affiliation(s)
- K Al-Tarrah
- Institute of Inflammation and Ageing, Birmingham University Medical School, B15 2TT, Birmingham, UK.,Scar Free Foundation Centre for Burns Research, University Hospital Birmingham Foundation Trust, B15 2WB, Birmingham, UK
| | - N Moiemen
- Scar Free Foundation Centre for Burns Research, University Hospital Birmingham Foundation Trust, B15 2WB, Birmingham, UK
| | - J M Lord
- Institute of Inflammation and Ageing, Birmingham University Medical School, B15 2TT, Birmingham, UK
| |
Collapse
|
9
|
Glassberg MK, Catanuto P, Shahzeidi S, Aliniazee M, Lilo S, Rubio GA, Elliot SJ. Estrogen deficiency promotes cigarette smoke-induced changes in the extracellular matrix in the lungs of aging female mice. Transl Res 2016; 178:107-117. [PMID: 27519148 DOI: 10.1016/j.trsl.2016.07.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/12/2016] [Accepted: 07/17/2016] [Indexed: 02/07/2023]
Abstract
Female smokers have a faster decline in lung function with increasing age and overall develop a greater loss of lung function than male smokers. This raises the question of whether estrogen status in women affects susceptibility to cigarette smoke (CS)-induced lung disease. Mouse models suggest that female mice are more susceptible than males to CS-induced lung disease. Moreover, young CS-exposed female mice develop emphysema earlier than male mice. The purpose of this study was to characterize the relationship of estrogen status on the pattern and severity of CS-induced lung disease. In this study, 15-month-old female C57BL/6J mice were ovariectomized and administered either placebo (pla) or 17β-estradiol (E2, 0.025 mg) 2 weeks after ovariectomy. They were further divided into those that were exposed to CS and no-smoke controls (NSC). Mice were exposed to CS in stainless steel inhalation chambers 3 hours a day, 5 days a week for 6 months, and sacrificed after 24 weeks of CS exposure. Blood and urine were collected at sacrifice to measure estrogen and cotinine levels, a metabolite of nicotine. Uterine weight was recorded as an indicator of estrogen status. Results showed that CS in the absence of E2 induced a decrease in hydroxyproline content, macrophage number, and respiratory chain complex-1 protein. CS without E2 also resulted in an increase in matrix metalloproteinase-2 activity and apoptosis and a change in the ratio of estrogen receptor subtype. These findings were abrogated with administration of E2, suggesting that estrogen deficiency increases susceptibility to CS-induced lung disease.
Collapse
Affiliation(s)
- Marilyn K Glassberg
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, Fla; Department of Surgery, Miller School of Medicine, University of Miami, Miami, Fla.
| | - Paola Catanuto
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Fla
| | - Shahriar Shahzeidi
- Division of Pediatric Pulmonology, Department of Pediatrics, Miller School of Medicine, University of Miami, Miami, Fla
| | | | - Sarit Lilo
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, Fla
| | - Gustavo A Rubio
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Fla
| | - Sharon J Elliot
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Fla
| |
Collapse
|
10
|
Chakraborty TR, Cohen J, Yohanan D, Alicea E, Weeks BS, Chakraborty S. Estrogen is neuroprotective against hypoglycemic injury in murine N38 hypothalamic cells. Mol Med Rep 2016; 14:5677-5684. [PMID: 27878271 DOI: 10.3892/mmr.2016.5952] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/25/2016] [Indexed: 11/05/2022] Open
Abstract
Estrogen (E2) has been demonstrated to possess protective effects from hypoglycemic toxicity, particularly in the pancreas. In the central nervous system, several brain regions, such as the hypothalamus, are highly vulnerable to hypoglycemic injuries that may lead to seizures, coma, and mortality. The present study performed a novel in vitro assay of hypoglycemic injury to hypothalamic cells, and is the first study, to the best of our knowledge, to demonstrate that E2 protects hypothalamic cells from hypoglycemic toxicity. The toxic effects of hypoglycemia on hypothalamic cells in vitro was determined by performing cell counts, together with MTT and lactate dehydrogenase assays, using the N38 murine hypothalamic cell line. Following 24 and 48 h in hypoglycemic conditions, a 60 and 75% reduction in cell number and mitochondrial function was observed, which reached 80 and ~100% by 72 and 96 h, respectively. E2 treatment prevented the hypoglycemia‑induced loss in cell number and mitochondrial toxicity at 24 and 48 h. However at 72 and 96 h of hypoglycemic conditions, the neuroprotective effects of E2 on cell number or mitochondrial function was not significant or not present at all. In order to determine whether E2 exerted its effects through the AKT signaling pathway, the expression of proline‑rich AKT substrate of 40 kDa (PRAS40) was analyzed. No alterations in PRAS40 expression were observed when N38 cells were exposed to hypoglycemic shock. From the biochemical and molecular data obtained, the authors speculated that E2 exhibits neuroprotective effects against hypoglycemic shock in hypothalamic cells, which dissipates with time. Despite demonstrating no significant effect on total AKT/PRS40 activity, it is possible that E2 may mediate these neuroprotective effects by upregulating the phosphorylated‑AKT/pPRAS40 signaling pathway. The present study presented, to the best of our knowledge, the first in vitro model for hypoglycemic toxicity to hypothalamic cells, and provided evidence to suggest that E2 may protect hypothalamic cells from the damaging effects of hypoglycemia.
Collapse
Affiliation(s)
| | - Joshua Cohen
- Department of Biology, Adelphi University, Garden City, NY 11530, USA
| | - Darien Yohanan
- Department of Biology, Adelphi University, Garden City, NY 11530, USA
| | - Eilliut Alicea
- Department of Biology, Adelphi University, Garden City, NY 11530, USA
| | - Benjamin S Weeks
- Department of Biology, Adelphi University, Garden City, NY 11530, USA
| | - Sanjoy Chakraborty
- Department of Biological Sciences, New York City College of Technology, City University of New York, New York, NY 11201, USA
| |
Collapse
|
11
|
Estrogen accelerates the resolution of inflammation in macrophagic cells. Sci Rep 2015; 5:15224. [PMID: 26477569 PMCID: PMC4609992 DOI: 10.1038/srep15224] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 09/17/2015] [Indexed: 12/15/2022] Open
Abstract
Although 17β-estradiol (E2) anti-inflammatory activity has been well described, very little is known about the effects of this hormone on the resolution phase of the inflammatory process. Here, we identified a previously unreported ERα-mediated effect of E2 on the inflammatory machinery. The study showed that the activation of the intracellular estrogen receptor shortens the LPS-induced pro-inflammatory phase and, by influencing the intrinsic and extrinsic programs, triggers the resolution of inflammation in RAW 264.7 cells. Through the regulation of the SOCS3 and STAT3 signaling pathways, E2 facilitates the progression of the inflammatory process toward the IL10-dependent “acquired deactivation” phenotype, which is responsible for tissue remodeling and the restoration of homeostatic conditions. The present study may provide an explanation for increased susceptibility to chronic inflammatory diseases in women after menopause, and it suggests novel anti-inflammatory treatments for such disorders.
Collapse
|
12
|
Mirzamohammadi S, Mehrabani M, Tekiyehmaroof N, Sharifi AM. Protective effect of 17β-estradiol on serum deprivation-induced apoptosis and oxidative stress in bone marrow-derived mesenchymal stem cells. Hum Exp Toxicol 2015; 35:312-22. [PMID: 25964380 DOI: 10.1177/0960327115586208] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Stem cell transplantation has indicated great promise for cell therapy in a wide range of diseases, but poor and insufficient viability of cells within damaged tissues has limited its potential therapeutic effects. 17 β-Estradiol (E2) is a steroid hormone that plays an important role in expression of many genes and regulating proliferation, viability, and intracellular redox status in different cell types. In this study, we aimed to assess the effect of E2 on bone marrow-derived mesenchymal stem cells (BM-MSCs). Apoptosis was induced by serum deprivation (SD), and cells were exposed to E2 in the presence or absence of serum for varying periods of time, after which cell viability was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Expression of proapoptotic and antiapoptotic proteins after exposure to E2 was examined by Western blotting. The ability of E2 to prevent reactive oxygen species (ROS) production was also measured. The results indicated that E2 significantly enhanced the viability of the cells and protected BM-MSCs against SD-induced overproduction of ROS. It could reduce lipid peroxidation, total antioxidant power, and also Bax/Bcl-2 ratio as well as expression of caspase-3. Taken together, our data support that E2 treatment protects BM-MSCs against SD-induced damage by regulating ROS production and upregulation of antiapoptotic/proapoptotic proteins ratio.
Collapse
Affiliation(s)
- S Mirzamohammadi
- Razi Drug Research Center, Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - M Mehrabani
- Razi Drug Research Center, Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - N Tekiyehmaroof
- Razi Drug Research Center, Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - A M Sharifi
- Razi Drug Research Center, Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Yehuda I, Madar Z, Leikin-Frenkel A, Tamir S. Glabridin, an isoflavan from licorice root, downregulates iNOS expression and activity under high-glucose stress and inflammation. Mol Nutr Food Res 2015; 59:1041-52. [PMID: 25737160 DOI: 10.1002/mnfr.201400876] [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] [Received: 11/30/2014] [Revised: 02/14/2015] [Accepted: 02/16/2015] [Indexed: 12/13/2022]
Abstract
SCOPE In females, hyperglycemia abolishes estrogen-vascular protection, leading to inflammation and oxidative stress that are related to diabetes-associated cardiovascular complications. Such knowledge led us to examine the potential of glabridin, as a replacement of estrogen anti-inflammatory activity under high-glucose conditions. METHODS AND RESULTS In macrophage-like cells, chronic glucose stress (28 and 44 mM) upregulated inducible nitric oxide synthase (iNOS) mRNA expression by 42 and 189%, respectively. Pretreatment with glabridin, under chronic glucose stress, downregulated the LPS-induced nitric oxide secretion and nitrotyrosine formation, by 39 and 21%, respectively. Pretreatment with estradiol did not prevent the LPS-induced nitrotyrosine formation. Furthermore, glabridin, brought about a decrease in the LPS-induced iNOS mRNA expression by 48%, as compared to cells pretreated with estradiol. Glabridin decreased protein levels of liver iNOS by 69% in adult mouse offspring which developed hyperglycemia after early fetal exposure to a saturated fatty acid-enriched maternal diet. Glabridin also decreased liver nitrotyrosine levels in offspring of regular diet-fed mothers after further receiving high-fat diet. CONCLUSION Such results indicate that glabridin retains anti-inflammatory abilities to regulate the synthesis and activity of iNOS under high-glucose levels, implying that a glabridin supplement may serve as an anti-inflammatory agent in diabetes-related vascular dysfunction.
Collapse
Affiliation(s)
- Itamar Yehuda
- Laboratory of Human Health and Nutrition Sciences, MIGAL-Galilee Research Institute, Kiryat-Shmona, Israel.,The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Jerusalem, Israel
| | - Zecharia Madar
- The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Jerusalem, Israel
| | - Alicia Leikin-Frenkel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Sheba Medical Center, Bert W. Strassburger Lipid Center, Tel-Hashomer, Israel
| | - Snait Tamir
- Laboratory of Human Health and Nutrition Sciences, MIGAL-Galilee Research Institute, Kiryat-Shmona, Israel.,Faculty of Sciences and Technology, Tel-Hai Academic College, Upper Galilee, Israel
| |
Collapse
|
14
|
Khusniyati E, Sari AA, Yueniwati Y, Noorhamdani N, Nurseta T, Keman K. The effects of Vigna unguiculata on cardiac oxidative stress and aorta estrogen receptor-β expression of ovariectomized rats. ASIAN PACIFIC JOURNAL OF REPRODUCTION 2014. [DOI: 10.1016/s2305-0500(14)60037-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
15
|
Fujihara M, Comizzoli P, Keefer CL, Wildt DE, Songsasen N. Epidermal Growth Factor (EGF) Sustains In Vitro Primordial Follicle Viability by Enhancing Stromal Cell Proliferation via MAPK and PI3K Pathways in the Prepubertal, but Not Adult, Cat Ovary1. Biol Reprod 2014; 90:86. [DOI: 10.1095/biolreprod.113.115089] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
16
|
Wang H, Guo Y, Zhao X, Li H, Fan G, Mao H, Miao L, Gao X. An estrogen receptor dependent mechanism of Oroxylin A in the repression of inflammatory response. PLoS One 2013; 8:e69555. [PMID: 23922737 PMCID: PMC3726624 DOI: 10.1371/journal.pone.0069555] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 06/10/2013] [Indexed: 11/17/2022] Open
Abstract
Oroxylin A, a natural flavonoid, is one of the main bioactive compounds that underlie the anti-inflammatory effect of the medicinal herb Scutellaria baicalensis Georgi widely used in southeastern Asia; however, the molecular mechanisms for the therapeutic benefits remain largely unclear. In this study, we found that Oroxylin A induces estrogen-responsive gene expression and promoter activity. In macrophages, Oroxylin A treatment significantly attenuates lipopolysaccharide (LPS)-induced but not basal inflammatory response, including nitric oxide (NO) production and the expression of inflammatory mediators (i.e., iNOS and COX-2) and cytokines (i.e., TNF-α, IL-1β, and IL-6), in an estrogen receptor (ER)-dependent manner. Oroxylin A treatment also dramatically decreases LPS-induced secretion of pro-inflammatory cytokines. Furthermore, the downregulation of all these inflammatory parameters by Oroxylin A was abolished when cells were pretreated with specific ER antagonist. Thus, Oroxylin A is a novel phytoestrogen and exhibits anti-inflammatory effects that are mediated by ER activity.
Collapse
Affiliation(s)
- Hong Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, China
| | - Ying Guo
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin Zhao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huiying Li
- Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, China
| | - Guanwei Fan
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Haoping Mao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Miao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiumei Gao
- Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
17
|
Role of sex steroid hormones in bacterial-host interactions. BIOMED RESEARCH INTERNATIONAL 2012; 2013:928290. [PMID: 23509808 PMCID: PMC3591248 DOI: 10.1155/2013/928290] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Accepted: 09/18/2012] [Indexed: 12/14/2022]
Abstract
Sex steroid hormones play important physiological roles in reproductive and nonreproductive tissues, including immune cells. These hormones exert their functions by binding to either specific intracellular receptors that act as ligand-dependent transcription factors or membrane receptors that stimulate several signal transduction pathways. The elevated susceptibility of males to bacterial infections can be related to the usually lower immune responses presented in males as compared to females. This dimorphic sex difference is mainly due to the differential modulation of the immune system by sex steroid hormones through the control of proinflammatory and anti-inflammatory cytokines expression, as well as Toll-like receptors (TLRs) expression and antibody production. Besides, sex hormones can also affect the metabolism, growth, or virulence of pathogenic bacteria. In turn, pathogenic, microbiota, and environmental bacteria are able to metabolize and degrade steroid hormones and their related compounds. All these data suggest that sex steroid hormones play a key role in the modulation of bacterial-host interactions.
Collapse
|
18
|
Xu Y, Fan H, Li X, Sun L, Hou Y. 17β-Estradiol enhances response of mice spleen B cells elicited by TLR9 agonist. Cell Immunol 2012; 278:125-35. [PMID: 22960279 DOI: 10.1016/j.cellimm.2012.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 06/27/2012] [Accepted: 07/13/2012] [Indexed: 12/23/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the production of autoantibodies against nucleic acid-associated antigens. B cells play cardinal roles in SLE. Many evidences have proved estrogen contribute to the gender bias in SLE and 17β-estradiol (E2) could accelerate the disease by regulating B cells. On the other hand, B cells express TLR9 which recognized dsDNA and played a critical role in SLE. However, the crosstalk between estrogen and TLR9 in B cells remains unknown. So we investigated the E2 effect in the presence of the TLR9 ligand CpG on mice spleen B cells. We found that the up-regulation of cell viability, life-span, co-stimulation molecules (CD40, CD86) expression, IgM secretion, TLR9 and MCM6 expression were more significant than CpG ODN or E2 stimulated alone. It may provide a new way to investigate the mechanism of how E2 modulate the B cells function in lupus.
Collapse
Affiliation(s)
- Yixin Xu
- Immunology and Reproductive Biology Lab, Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, PR China
| | | | | | | | | |
Collapse
|
19
|
Faroni A, Magnaghi V. The neurosteroid allopregnanolone modulates specific functions in central and peripheral glial cells. Front Endocrinol (Lausanne) 2011; 2:103. [PMID: 22654838 PMCID: PMC3356145 DOI: 10.3389/fendo.2011.00103] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Accepted: 12/05/2011] [Indexed: 12/15/2022] Open
Abstract
Since the first observations on the existence of "neurosteroids" in the 1980s, our understanding of the importance of these endogenous steroids in the control of the central and peripheral nervous system (PNS) has increased progressively. Although most of the observations were made in neuronal cells, equally important are the effects that neurosteroids exert on glial cells. Among the different classes of neurosteroids acting on glial cells, the progesterone 5α-3α metabolite, allopregnanolone, displays a particular mechanism of action involving primarily the modulation of classic GABA receptors. In this review, we focus our attention on allopregnanolone because its effects on the physiology of glial cells of the central and PNS are intriguing and could potentially lead to the development of new strategies for neuroprotection and/or regeneration of injured nervous tissues.
Collapse
Affiliation(s)
- Alessandro Faroni
- Blond McIndoe Laboratories, Regenerative Biomedicine, School of Medicine, The University of ManchesterManchester, UK
- Department of Endocrinology, Physiopathology, Applied Biology, University of MilanMilan, Italy
| | - Valerio Magnaghi
- Department of Endocrinology, Physiopathology, Applied Biology, University of MilanMilan, Italy
- *Correspondence: Valerio Magnaghi, Department of Endocrinology, Physiopathology, Applied Biology, University of Milan, Via G. Balzaretti 9, 20133 Milan, Italy. e-mail:
| |
Collapse
|
20
|
Park G, Jeong JW, Kim JE. SIRT1 deficiency attenuates MPP+-induced apoptosis in dopaminergic cells. FEBS Lett 2010; 585:219-24. [DOI: 10.1016/j.febslet.2010.11.048] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Revised: 11/13/2010] [Accepted: 11/25/2010] [Indexed: 01/28/2023]
|
21
|
Winzer M, Rauner M, Pietschmann P. Glycitein decreases the generation of murine osteoclasts and increases apoptosis. Wien Med Wochenschr 2010; 160:446-51. [DOI: 10.1007/s10354-010-0811-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Accepted: 03/21/2010] [Indexed: 10/19/2022]
|
22
|
Subramanian M, Shaha C. Oestrogen modulates human macrophage apoptosis via differential signalling through oestrogen receptor-alpha and beta. J Cell Mol Med 2010. [PMID: 20141615 DOI: 10.1111/j.1582-4934.2008.00679.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human macrophages express oestrogen receptors and are therefore competent to respond to the hormone present in their microenvironment, which is implicated in sexual dimorphism observed in several immune and autoimmune phenomena. An earlier study from this laboratory demonstrated 17beta-oestradiol (E2) induced apoptosis in macrophages derived from human peripheral blood monocytes and THP-1 acute monocytic leukaemia cell line when Bcl-2 was down-regulated; however, the involvement of E2 receptor subtypes in the modulation of death pathways in these cells remain unknown. Using macrophages derived from THP-1 human acute monocytic leukaemia cells as a model, we demonstrate that plasma membrane associated oestrogen receptor (ER) -alpha participate in E2 induced Bcl-2 increase, through activation of the mitogen activated protein kinase (MAPK) pathway whereas cytosolic ER-beta transmits signals for the pro-apoptotic event of Bax translocation. The mechanistic basis of Bax translocation comprised of ER-beta mediated increase in intracellular pH, facilitated by activation of the Na(+)-H(+) exchanger. Intracellular alkalinization accompanied by concomitant Bcl-2 increase and Bax migration does not cause cellular apoptosis; however, siRNA mediated down-regulation of ER-alpha during E2 exposure leads to inhibition of Bcl-2 increase and consequently apoptosis due to the unopposed action of mitochondrial Bax. In summary, this study underscores the importance of integrative signalling modality from multiple oestrogen receptor pools in modulating oestrogen effects on human monocyte-derived macrophage apoptotic signalling pathway, which opens new vistas to explore the use of selective oestrogen receptor modulators in apoptosis-based therapies.
Collapse
Affiliation(s)
- Manikandan Subramanian
- Cell Death and Differentiation Research Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | | |
Collapse
|
23
|
Nadzialek S, Pigneur LM, Wéron B, Kestemont P. Bcl-2 and Caspase 3 mRNA levels in the testes of gudgeon, Gobio gobio, exposed to ethinylestradiol (EE2). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2010; 98:304-310. [PMID: 20356636 DOI: 10.1016/j.aquatox.2010.02.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2009] [Revised: 02/19/2010] [Accepted: 02/23/2010] [Indexed: 05/29/2023]
Abstract
Apoptosis inhibition has been reported in the male reproductive tract of teleost fish exposed to 17beta-estrogen or estrogen-like compounds. In order to understand the molecular mechanisms of cell death inhibition, this study examined 2 genes involved in the apoptotic pathway, Bcl-2 and Caspase 3, an anti-apoptotic and a pro-apoptotic genes, respectively. Partial cDNA sequences of Bcl-2 and Caspase 3 were cloned from gudgeon (Gobio gobio), a common European cyprinid fish. To follow mRNA levels of Bcl-2 and Caspase 3 under xenoestrogen exposure, we first performed an in vitro experiment on fish testis exposed to the most potent xenoestrogen found in the environment, ethinylestradiol (EE2). We further studied mRNA expression of both genes in the testis of fish exposed to xenoestrogens in situ. In the in vitro experiment, fragments of gudgeon testis were exposed for 21 days to 10(-3), 10(-2), 10(-1), 1 and 10 microg/L of EE2, as well as to positive (10(-1) microg/L of E2) and ethanol control medium. Results showed a significant induction of Bcl-2 mRNA at 10(-1) microg/L (p<0.05). Surprisingly, Caspase 3, a cell death effector, displayed the same profile as observed for the anti-apoptotic gene Bcl-2. In the experiment on wild gudgeon exposed from birth to an estrogenic sewage treatment plant effluent, the mRNA expression of Bcl-2 and Caspase 3 in feminized fish (ovotestis) was not significantly different due to high variability of expression between individuals. At the current state of knowledge on spermatogenesis disruption in teleost fish, in vitro studies seem better adapted than in situ investigations to enlighten the molecular pathway of apoptosis inhibition in testis exposed to xenoestrogens.
Collapse
Affiliation(s)
- Stéphanie Nadzialek
- The University of Namur (FUNDP), Unité de Recherche en Biologie des Organismes (URBO), 61 rue de Bruxelles, B-5000 Namur, Belgium
| | - Lise-Marie Pigneur
- The University of Namur (FUNDP), Unité de Recherche en Biologie des Organismes (URBO), 61 rue de Bruxelles, B-5000 Namur, Belgium
| | - Benjamin Wéron
- The University of Namur (FUNDP), Unité de Recherche en Biologie des Organismes (URBO), 61 rue de Bruxelles, B-5000 Namur, Belgium
| | - Patrick Kestemont
- The University of Namur (FUNDP), Unité de Recherche en Biologie des Organismes (URBO), 61 rue de Bruxelles, B-5000 Namur, Belgium.
| |
Collapse
|
24
|
Kyurkchiev D, Ivanova-Todorova E, Kyurkchiev SD. New target cells of the immunomodulatory effects of progesterone. Reprod Biomed Online 2010; 21:304-11. [PMID: 20638907 DOI: 10.1016/j.rbmo.2010.04.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 04/01/2010] [Accepted: 04/07/2010] [Indexed: 01/08/2023]
Abstract
It is well known that the reproductive steroid hormones, particularly progesterone, in addition to its widely recognized effects on endometrial epithelial and stromal cells and spiral arteries, affect the activities of T cells and natural killer cells in the deciduas, thus inducing active immune tolerance against the fetal antigens. The immunomodulatory effects of progesterone on T cells, B cells and natural killer cells have been discussed extensively in the literature. The aim of the present review is to sum up and discuss the results from this and other laboratories of investigations on the effects of progesterone on dendritic cells and adult stem cells, which are some of the other cell populations present at the fetal-maternal interface and possibly are related to the immunoregulation during pregnancy. These cells have been shown to have a number of specific functions but their involvement in the entire process of regulation of the immune response in pregnancy is still under discussion. The present review focuses on facts showing that the progesterone is a kind of 'regulator of regulators' in the decidua, thus creating the most favourable conditions for the development of the semi-allogeneic fetus in successful pregnancy.
Collapse
Affiliation(s)
- Dobroslav Kyurkchiev
- Laboratory of Clinical Immunology, University Hospital St. Ivan Rilski, Medical University Sofia, 15 Acad. Ivan Geshov, 1431 Sofia, Bulgaria.
| | | | | |
Collapse
|
25
|
Nadzialek S, Kestemont P. 17alpha-ethinylestradiol induces an imbalance between apoptosis and cell proliferation to sex steroid disruption in a testis culture of gudgeon, Gobio gobio. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2010; 29:881-886. [PMID: 20821517 DOI: 10.1002/etc.100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
The aim of this study was to investigate the effect of the most potent xenoestrogen currently found in the environment, ethinylestradiol (EE2), on some physiological events occurring during early spermatogenesis of gudgeon (Gobio gobio), a common European fish species. Physiological pathways studies were apoptosis, cell proliferation, and steroidogenesis on sex steroids (testosterone [T], 11-ketotestosterone [11-KT], and 17beta-estradiol [E2]). Testis pieces were cultured in vitro during 21 d at 10(-4), 10(-3), 10(-2), 10(-1), 1 and 10 microg/L of EE2 as well as in positive (10(-1) microg/L of E2) and ethanol control medium. Apoptosis and cell proliferation displayed opposite responses related to the EE2 concentration. When apoptosis inhibition was observed, cell proliferation was induced at 10(-2) and 10(-1) microg/L of EE2 as well as in the positive control. In contrast, a massive cell death was detected for high EE2 concentrations (1 and 10 microg/L). Steroidogenesis was also disrupted in a dose-related manner. 11-Ketotestosterone was depressed at 10(-2) and 10(-1) microg/L of EE2 whereas E2 was detectable in the medium only at 10(-3), 10(-2), and 10(-1) microg/L of EE2. High concentrations of T were detected in the medium at 10(-3), 10(-2), and 10(-1) microg/L of EE2 but depressed at 1 and 10 microg/L of EE2. In conclusion, intermediate EE2 concentrations (10(-2) and 10(-1) microg/L) used in this experimental design have obviously disrupted early spermatogenesis, leading to an imbalance between cell death and cell proliferation in a sex steroid environment toward E2. The results of the present study could be the basis conditions for oocyte development within the testis of a common teleost fish under xenoestrogen exposure.
Collapse
Affiliation(s)
- Stéphanie Nadzialek
- Unité de Recherche en Biologie des Organismes (URBO), University of Namur (FUNDP), 61 rue de Bruxelles, 5000 Namur, Belgium
| | | |
Collapse
|
26
|
Crisafulli C, Bruscoli S, Esposito E, Mazzon E, Di Paola R, Genovese T, Bramanti P, Migliorati G, Cuzzocrea S. PPAR-alpha contributes to the anti-inflammatory activity of 17beta-estradiol. J Pharmacol Exp Ther 2009; 331:796-807. [PMID: 19755663 DOI: 10.1124/jpet.109.156646] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Because studies have shown that 17beta-estradiol (E2) produces anti-inflammatory effects after various adverse circulatory conditions, we have recently demonstrated that E2 significantly reduced the acute lung injury. Moreover, previous results suggest that peroxisome proliferator-activated receptor-alpha (PPAR-alpha), an intracellular transcription factor activated by fatty acids, plays a role in the control of inflammation. With the aim to characterize the role of PPAR-alpha in estrogen-mediated anti-inflammatory activity, we tested the efficacy of E2 in an experimental model of lung inflammation, carrageenan-induced pleurisy, comparing ovariectomized wild-type (WT) and PPAR-alpha lacking (PPAR-alphaKO) mice. Results indicate that E2-mediated anti-inflammatory activity is weakened in PPAR-alphaKO mice, compared with WT control groups. In particular, E2 was less effective in PPAR-alphaKO, compared with WT mice, in inhibition of cell migration as well as lung injury, NF-kB activation, TNF-alpha production, and inducible nitric-oxide synthase (iNOS) activation. Moreover, macrophages from PPAR-alphaKO were less susceptible to E2-induced iNOS inhibition in vitro compared with macrophages from WT mice. Moreover, the results indicate that PPAR-alpha was required for estrogen receptor up-regulation, following E2 treatment. These results show for the first time that PPAR-alpha contributes to the anti-inflammatory activity of E2.
Collapse
Affiliation(s)
- Concetta Crisafulli
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, 98125 Messina, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Collier FM, Loving A, Baker AJ, McLeod J, Walder K, Kirkland MA. RTKN2 Induces NF-KappaB Dependent Resistance to Intrinsic Apoptosis in HEK Cells and Regulates BCL-2 Genes in Human CD4(+) Lymphocytes. J Cell Death 2009; 2:9-23. [PMID: 26124677 PMCID: PMC4474337 DOI: 10.4137/jcd.s2891] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The gene for Rhotekin 2 (RTKN2) was originally identified in a promyelocytic cell line resistant to oxysterol-induced apoptosis. It is differentially expressed in freshly isolated CD4+ T-cells compared with other hematopoietic cells and is down-regulated following activation of the T-cell receptor. However, very little is known about the function of RTKN2 other than its homology to Rho-GTPase effector, rhotekin, and the possibility that they may have similar roles. Here we show that stable expression of RTKN2 in HEK cells enhanced survival in response to intrinsic apoptotic agents; 25-hydroxy cholesterol and camptothecin, but not the extrinsic agent, TNFα. Inhibitors of NF-KappaB, but not MAPK, reversed the resistance and mitochondrial pro-apoptotic genes, Bax and Bim, were down regulated. In these cells, there was no evidence of RTKN2 binding to the GTPases, RhoA or Rac2. Consistent with the role of RTKN2 in HEK over-expressing cells, suppression of RTKN2 in primary human CD4+ T-cells reduced viability and increased sensitivity to 25-OHC. The expression of the pro-apoptotic genes, Bax and Bim were increased while BCL-2 was decreased. In both cell models RTKN2 played a role in the process of intrinsic apoptosis and this was dependent on either NF-KappaB signaling or expression of downstream BCL-2 genes. As RTKN2 is a highly expressed in CD4+ T-cells it may play a role as a key signaling switch for regulation of genes involved in T-cell survival.
Collapse
Affiliation(s)
- Fiona M Collier
- Barwon Biomedical Research, Geelong Hospital, Barwon Health, Ryrie St, Geelong, Victoria, 3227, Australia. ; Metabolic Research Unit, School of Medicine and Institute for Technology Research and Innovation, Deakin University, Waurn Ponds, Victoria, 3217, Australia
| | - Andrea Loving
- Barwon Biomedical Research, Geelong Hospital, Barwon Health, Ryrie St, Geelong, Victoria, 3227, Australia
| | - Adele J Baker
- Department of Malignant Haematology and Stem Cell Transplantation, The Alfred Hospital, Melbourne, Victoria, 3181, Australia
| | - Janet McLeod
- School of Medicine, Deakin University, Waurn Ponds, Victoria, 3217, Australia
| | - Ken Walder
- Metabolic Research Unit, School of Medicine and Institute for Technology Research and Innovation, Deakin University, Waurn Ponds, Victoria, 3217, Australia
| | - Mark A Kirkland
- Barwon Biomedical Research, Geelong Hospital, Barwon Health, Ryrie St, Geelong, Victoria, 3227, Australia
| |
Collapse
|
28
|
Maselli A, Matarrese P, Straface E, Canu S, Franconi F, Malorni W. Cell sex: a new look at cell fate studies. FASEB J 2009; 23:978-84. [PMID: 19074513 DOI: 10.1096/fj.08-114348] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2025]
Abstract
Cell death processes have been widely investigated in recent years in order to elucidate the different pathways involved in the complex machinery implicated in determining cell fate. Different forms of cell death have been described: Apart from the classical form of death known as necrosis, a well-characterized traumatic injury of the cell, several additional forms of cell death have been identified. Of these, apoptosis has been characterized in the greatest detail. Defects in the mechanisms of cell demise (that is, an excess of or decrease in apoptosis) have been associated with the pathogenesis of a number of human diseases. Here we review some new aspects derived from recent insights into this field, particularly the hypothesis that cells of males and females could display several different features, including those determining their fate.
Collapse
Affiliation(s)
- Angela Maselli
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | |
Collapse
|
29
|
Brufani M, Ceccacci F, Filocamo L, Garofalo B, Joudioux R, La Bella A, Leonelli F, Migneco LM, Marini Bettolo R, Farina PM, Ashcroft GS, Routley C, Hardman M, Meda C, Rando G, Maggi A. Novel Locally Active Estrogens Accelerate Cutaneous Wound Healing. A Preliminary Study. Mol Pharm 2009; 6:543-56. [DOI: 10.1021/mp800206b] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Mario Brufani
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Francesca Ceccacci
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Luigi Filocamo
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Barbara Garofalo
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Roberta Joudioux
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Angela La Bella
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Francesca Leonelli
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Luisa M. Migneco
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Rinaldo Marini Bettolo
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Paolo M. Farina
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Gillian S. Ashcroft
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Claire Routley
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Matthew Hardman
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Clara Meda
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Gianpaolo Rando
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| | - Adriana Maggi
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Università degli Studi di Roma “La Sapienza”, via degli Apuli 9, I-00185 Roma, Italy, Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma “La Sapienza”, P.le Aldo Moro 5, I-00185 Roma, Italy, Euticals Ambrosia S.p.A., Via Monte Rosa, 114/116, 20089 Rozzano (MI), Italy, Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, U.K.,
| |
Collapse
|
30
|
Vegeto E, Benedusi V, Maggi A. Estrogen anti-inflammatory activity in brain: a therapeutic opportunity for menopause and neurodegenerative diseases. Front Neuroendocrinol 2008; 29:507-19. [PMID: 18522863 PMCID: PMC2630539 DOI: 10.1016/j.yfrne.2008.04.001] [Citation(s) in RCA: 237] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Revised: 04/18/2008] [Accepted: 04/21/2008] [Indexed: 11/28/2022]
Abstract
Recent studies highlight the prominent role played by estrogens in protecting the central nervous system (CNS) against the noxious consequences of a chronic inflammatory reaction. The neurodegenerative process of several CNS diseases, including Multiple Sclerosis, Alzheimer's and Parkinson's Diseases, is associated with the activation of microglia cells, which drive the resident inflammatory response. Chronically stimulated during neurodegeneration, microglia cells are thought to provide detrimental effects on surrounding neurons. The inhibitory activity of estrogens on neuroinflammation and specifically on microglia might thus be considered as a beneficial therapeutic opportunity for delaying the onset or progression of neurodegenerative diseases; in addition, understanding the peculiar activity of this female hormone on inflammatory signalling pathways will possibly lead to the development of selected anti-inflammatory molecules. This review summarises the evidence for the involvement of microglia in neuroinflammation and the anti-inflammatory activity played by estrogens specifically in microglia.
Collapse
Affiliation(s)
- Elisabetta Vegeto
- Centre of Excellence on Neurodegenerative Disease, University of Milan via Balzaretti, 9 20133-Milan, Italy
| | - Valeria Benedusi
- Centre of Excellence on Neurodegenerative Disease, University of Milan via Balzaretti, 9 20133-Milan, Italy
| | - Adriana Maggi
- Centre of Excellence on Neurodegenerative Disease, University of Milan via Balzaretti, 9 20133-Milan, Italy
| |
Collapse
|
31
|
Studies of the effects of new progestins from the pentarane series on viability, epidermal growth factor receptor expression, and apoptosis of cultured HeLa and MCF-7 cells. Pharm Chem J 2008. [DOI: 10.1007/s11094-008-0102-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
32
|
Kyurkchiev D, Ivanova-Todorova E, Hayrabedyan S, Altankova I, Kyurkchiev S. Female Sex Steroid Hormones Modify Some Regulatory Properties of Monocyte-Derived Dendritic Cells. Am J Reprod Immunol 2007; 58:425-33. [DOI: 10.1111/j.1600-0897.2007.00526.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
33
|
Malorni W, Campesi I, Straface E, Vella S, Franconi F. Redox features of the cell: a gender perspective. Antioxid Redox Signal 2007; 9:1779-801. [PMID: 17822369 DOI: 10.1089/ars.2007.1596] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Reactive oxygen and nitrogen species have been implicated in diverse subcellular activities, including cell proliferation,differentiation and, in some instances, cell injury and death. The implications of reactive species inhuman pathology have also been studied in detail. However, although the role of free radicals in the pathogenesis of human diseases has been extensively analyzed in different systems (i.e., in vitro, ex vivo, and in vivo),it is still far from elucidated. In particular, the possible role of gender 4 differences in human pathophysiology associated with reactive species is a promising new field of investigation. Although the complex scenario this presents is still incomplete, important gender-associated "redox features" of cells have already been described in the literature. Here we summarize the different aspects of redox-associated molecules and enzymes in regard to gender differences in terms of the intracellular production and biochemical activity of reactive species. These are often associated with the pathogenetic mechanisms underlying several human morbidities(e.g., degenerative diseases) and can represent a specific target for new pharmacologic strategies. Gender differences may thus pose an important challenge for future studies aimed at the clinical management of diseases characterized by a redox imbalance.
Collapse
Affiliation(s)
- Walter Malorni
- Department of Drug Research and Evaluation, Istituto Superiore di Sanita', Rome, Italy.
| | | | | | | | | |
Collapse
|
34
|
Subramanian M, Shaha C. Up-regulation of Bcl-2 through ERK phosphorylation is associated with human macrophage survival in an estrogen microenvironment. THE JOURNAL OF IMMUNOLOGY 2007; 179:2330-8. [PMID: 17675494 DOI: 10.4049/jimmunol.179.4.2330] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Estrogen is a known immunomodulator with pleiotropic effects on macrophage function that partly accounts for the gender bias observed in numerous autoimmune, cardiovascular, and neurodegenerative disorders. The effect of estrogen on the survival of human macrophages is largely unknown, and in this study we demonstrate that 17beta-estradiol (E2) provokes a death response in human THP-1 macrophages by initiating Bax translocation from cytosol to the mitochondria; however, a concomitant up-regulation of Bcl-2 creates a Bax to Bcl-2 ratio favorable for Bcl-2, thus ensuring cell survival. Both Bcl-2 up-regulation and Bax translocation are estrogen receptor-dependent events; however, Bcl-2 augmentation but not Bax translocation is dependent on Ca(2+) increase, activation of protein kinase C, and ERK phosphorylation. This estrogen-induced Bcl-2 increase is crucial for the survival of THP-1 macrophages as well as that of human peripheral blood monocyte-derived macrophages, which is evident from E2-induced cell death under small interfering RNA-mediated Bcl-2 knockdown conditions. Hence, this study demonstrates that E2-induced Bcl-2 up-regulation is a homeostatic survival mechanism necessary for the manifestation of immunomodulatory effect of estrogen on human macrophages.
Collapse
|
35
|
Abstract
There is still an unresolved paradox with respect to the immunomodulating role of estrogens. On one side, we recognize inhibition of bone resorption and suppression of inflammation in several animal models of chronic inflammatory diseases. On the other hand, we realize the immunosupportive role of estrogens in trauma/sepsis and the proinflammatory effects in some chronic autoimmune diseases in humans. This review examines possible causes for this paradox. This review delineates how the effects of estrogens are dependent on criteria such as: 1) the immune stimulus (foreign antigens or autoantigens) and subsequent antigen-specific immune responses (e.g., T cell inhibited by estrogens vs. activation of B cell); 2) the cell types involved during different phases of the disease; 3) the target organ with its specific microenvironment; 4) timing of 17beta-estradiol administration in relation to the disease course (and the reproductive status of a woman); 5) the concentration of estrogens; 6) the variability in expression of estrogen receptor alpha and beta depending on the microenvironment and the cell type; and 7) intracellular metabolism of estrogens leading to important biologically active metabolites with quite different anti- and proinflammatory function. Also mentioned are systemic supersystems such as the hypothalamic-pituitary-adrenal axis, the sensory nervous system, and the sympathetic nervous system and how they are influenced by estrogens. This review reinforces the concept that estrogens have antiinflammatory but also proinflammatory roles depending on above-mentioned criteria. It also explains that a uniform concept as to the action of estrogens cannot be found for all inflammatory diseases due to the enormous variable responses of immune and repair systems.
Collapse
Affiliation(s)
- Rainer H Straub
- Laboratory of Experimental Rheumatology and Neuroendocrino-Immunology, Division of Rheumatology, Department of Internal Medicine I, University Hospital, 93042 Regensburg, Germany.
| |
Collapse
|
36
|
Abstract
Hormonal and locally produced steroids act in the nervous system as neuroendocrine regulators, as trophic factors and as neuromodulators and have a major impact on neural development and function. Glial cells play a prominent role in the local production of steroids and in the mediation of steroid effects on neurons and other glial cells. In this review, we examine the role of glia in the synthesis and metabolism of steroids and the functional implications of glial steroidogenesis. We analyze the mechanisms of steroid signaling on glia, including the role of nuclear receptors and the mechanisms of membrane and cytoplasmic signaling mediated by changes in intracellular calcium levels and activation of signaling kinases. Effects of steroids on functional parameters of glia, such as proliferation, myelin formation, metabolism, cytoskeletal reorganization, and gliosis are also reviewed, as well as the implications of steroid actions on glia for the regulation of synaptic function and connectivity, the regulation of neuroendocrine events, and the response of neural tissue to injury.
Collapse
|
37
|
Pozzi S, Benedusi V, Maggi A, Vegeto E. Estrogen action in neuroprotection and brain inflammation. Ann N Y Acad Sci 2006; 1089:302-23. [PMID: 17261778 DOI: 10.1196/annals.1386.035] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The fertile period of women's life compared to menopause is associated with a lower incidence of degenerative inflammatory diseases. In brain, estrogens ameliorate brain performance and have positive effects on selected neural pathologies characterized by a strong inflammatory component. We thus hypothesized that the inflammatory response is a target of estrogen action; several studies including ours provided strong evidence to support this prediction. Microglia, the brain's inflammatory cells, and circulating monocytes express the estrogen receptors ER-alpha and ER-beta and their responsiveness in vivo and in vitro to pro-inflammatory agents, such as lipopolysaccharide (LPS), is controlled by 17beta-estradiol (E(2)). Susceptibility of central nervous system (CNS) macrophage cells to E(2) is also preserved in animal models of neuroinflammatory diseases, in which ER-alpha seems to be specifically involved. At the molecular level, induction of inflammatory gene expression is blocked by E(2). We recently observed that, differently from conventional anti-inflammatory drugs, E(2) stimulates a nongenomic event that interferes with the LPS signal transduction from the plasma membrane to cytoskeleton and intracellular effectors, which results in the inhibition of the nuclear translocation of NF-kappaB, a transcription factor of inflammatory genes. Interference with NF-kappaB intracellular trafficking is selectively mediated by ER-alpha. In summary, evidence from basic research strongly indicates that the use of estrogenic drugs that can mimic the anti-inflammatory activity of E(2) might trigger beneficial effects against neurodegeneration in addition to carrying out their specific therapeutic function.
Collapse
Affiliation(s)
- Silvia Pozzi
- Center of Excellence on Neurodegenerative Diseases, Department of Pharmacological Sciences, University of Milan, Via Balzaretti, 9, 20133 Milan, Italy
| | | | | | | |
Collapse
|
38
|
Mourdjeva M, Popova Z, Kyurkchiev D, Kontinopoulou K, Altankova I, Kehayov I, Kyurkchiev S. Progesterone-Modulated Phosphatidylserine Externalization in Apoptosis and Activation of Jurkat Cells. Am J Reprod Immunol 2006; 56:249-57. [PMID: 16938114 DOI: 10.1111/j.1600-0897.2006.00422.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
PROBLEM During pregnancy the elevated levels of progesterone (Pg) have immunomodulating effects. It is important to follow-up Pg effects on basic biological processes at cell level as apoptosis and activation which was the aim of this study. METHODS OF STUDY Jurkat cells cultured in the presence or absence of Pg were used as a model system. Apoptosis was induced by H(2)O(2) and activation by phorbol myriastate acetate. The induced changes in the phosphatidylserine (PS) externalization and cell surface CD69 expression were followed by fluorescence-activated cell sorter and immunofluorescence. RESULTS After the induction of apoptosis PS externalizes in 52.3% of Jurkat cells. Cells cultured with Pg show tendency to a decrease of PS positive cells (42%). The opposite effect is observed in activated cells--PS externalization increase from 33.8% of control cells to 40.1% of Pg-treated cells. CONCLUSIONS These findings would suggest that by increasing activation and decreasing apoptosis Pg could regulate local immune system during pregnancy.
Collapse
Affiliation(s)
- Milena Mourdjeva
- Department of Molecular Immunology, Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, 73 Tzarigradsko shosse, 1113 Sofia, Bulgaria.
| | | | | | | | | | | | | |
Collapse
|
39
|
Williams TA, Verhovez A, Milan A, Veglio F, Mulatero P. Protective effect of spironolactone on endothelial cell apoptosis. Endocrinology 2006; 147:2496-505. [PMID: 16497808 DOI: 10.1210/en.2005-1318] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human umbilical vein endothelial cells (HUVECs) undergo apoptosis in response to serum deprivation. We show that the nonspecific mineralocorticoid receptor antagonist, spironolactone, protects from caspase-3 activation induced by serum deprivation in contrast to the selective mineralocorticoid receptor antagonist, eplerenone, that is nonprotective. We also demonstrate that progesterone, hydrocortisone, and dexamethasone all protect HUVECs from serum-deprivation-induced caspase-3 activation, whereas aldosterone and dihydrotestosterone have no effect. Spironolactone has been demonstrated to display agonist activity only to the progesterone receptor (PR), and we additionally show that spironolactone and progesterone, but not eplerenone, inhibit mitochondrial cytochrome c release and cleavage of nuclear poly (ADP-ribose) polymerase (PARP) and increase cell viability. Additionally, the PR antagonist mifepristone (RU486) partially blocked the inhibitory effect of both spironolactone and progesterone on caspase-3 activation, cytochrome c release, and nuclear PARP cleavage. Nitric oxide (NO) protects HUVECs from apoptosis in response to various stimuli including serum-deprivation; however, the NO synthase inhibitor N-monomethyl-l-arginine, did not abolish inhibition of caspase-3 activation or PARP cleavage by spironolactone. Thus, we demonstrate that spironolactone protects HUVECs from serum-deprivation-induced apoptosis by inhibition of caspase-3 activity, cytochrome c release and PARP cleavage by a NO-independent mechanism; further, this effect is likely mediated by the agonist properties of spironolactone toward the PR.
Collapse
Affiliation(s)
- Tracy A Williams
- Department of Medicine and Experimental Oncology, Hypertension Unit, University of Torino, Italy.
| | | | | | | | | |
Collapse
|
40
|
Vegeto E, Belcredito S, Ghisletti S, Meda C, Etteri S, Maggi A. The endogenous estrogen status regulates microglia reactivity in animal models of neuroinflammation. Endocrinology 2006; 147:2263-72. [PMID: 16469811 DOI: 10.1210/en.2005-1330] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It has been previously demonstrated that 17beta-estradiol (E(2)) inhibits the response of microglia, the resident brain macrophages, to acute injuries in specific brain regions. We here show that the effect of E(2) in acute brain inflammation is widespread and that the hormone reduces the expression of inflammatory mediators, such as monocyte chemoattractant protein-1, macrophage inflammatory protein-2, and TNF-alpha, induced by lipopolysaccharide, demonstrating that microglia are a direct target of estrogen action in brain. Using the APP23 mice, an animal model of Alzheimer's disease reproducing chronic neuroinflammation, we demonstrate that ovary ablation increases microglia activation at beta-amyloid (Abeta) deposits and facilitates the progression of these cells toward a highly reactive state. Long-term administration of E(2) reverts the effects of ovariectomy and decreases microglia reactivity compared with control animals. In this animal model, these events do not correlate with a reduced number of Abeta deposits. Finally, we show that E(2) inhibits Abeta-induced expression of scavenger receptor-A in macrophage cells, providing a mechanism for the effect of E(2) on Abeta signaling observed in the APP23 mice. Altogether, our observations reveal a substantial involvement of endogenous estrogen in neuroinflammatory processes and provide novel mechanisms for hormone action in the brain.
Collapse
Affiliation(s)
- Elisabetta Vegeto
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological Sciences, University of Milan, Italy.
| | | | | | | | | | | |
Collapse
|
41
|
Bryant DN, Sheldahl LC, Marriott LK, Shapiro RA, Dorsa DM. Multiple pathways transmit neuroprotective effects of gonadal steroids. Endocrine 2006; 29:199-207. [PMID: 16785596 DOI: 10.1385/endo:29:2:199] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Revised: 11/30/1999] [Accepted: 10/25/2005] [Indexed: 12/27/2022]
Abstract
Numerous preclinical studies suggest that gonadal steroids, particularly estrogen, may be neuroprotective against insult or disease progression. This paper reviews the mechanisms contributing to estrogen-mediated neuroprotection. Rapid signaling pathways, such as MAPK, PI3K, Akt, and PKC, are required for estrogen's ability to provide neuroprotection. These rapid signaling pathways converge on genomic pathways to modulate transcription of E2-responsive genes via ERE-dependent and ERE-independent mechanisms. It is clear that both rapid signaling and transcription are important for estrogen's neuroprotective effects. A mechanistic understanding of estrogen-mediated neuroprotection is crucial for the development of therapeutic interventions that enhance quality of life without deleterious side effects.
Collapse
Affiliation(s)
- Damani N Bryant
- Department of Physiology and Pharmacology (L334), Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | | | |
Collapse
|
42
|
Machida T, Fujita T, Ooo ML, Ohira M, Isogai E, Mihara M, Hirato J, Tomotsune D, Hirata T, Fujimori M, Adachi W, Nakagawara A. Increased expression of proapoptotic BMCC1, a novel gene with the BNIP2 and Cdc42GAP homology (BCH) domain, is associated with favorable prognosis in human neuroblastomas. Oncogene 2006; 25:1931-42. [PMID: 16288218 DOI: 10.1038/sj.onc.1209225] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Differential screening of the genes obtained from cDNA libraries of primary neuroblastomas (NBLs) between the favorable and unfavorable subsets has identified a novel gene BCH motif-containing molecule at the carboxyl terminal region 1 (BMCC1). Its 350 kDa protein product possessed a Bcl2-/adenovirus E1B nineteen kDa-interacting protein 2 (BNIP2) and Cdc42GAP homology domain in the COOH-terminus in addition to P-loop and a coiled-coil region near the NH2-terminus. High levels of BMCC1 expression were detected in the human nervous system as well as spinal cord, brain and dorsal root ganglion in mouse embryo. The immunohistochemical study revealed that BMCC1 was positively stained in the cytoplasm of favorable NBL cells but not in unfavorable ones with MYCN amplification. The quantitative real-time reverse transcription-PCR using 98 primary NBLs showed that high expression of BMCC1 was a significant indicator of favorable NBL. In primary culture of newborn mice superior cervical ganglion (SCG) neurons, mBMCC1 expression was downregulated after nerve growth factor (NGF)-induced differentiation, and upregulated during the NGF-depletion-induced apoptosis. Furthermore, the proapoptotic function of BMCC1 was also suggested by increased expression in CHP134 NBL cells undergoing apoptosis after treatment with retinoic acid, and by an enhanced apoptosis after depletion of NGF in the SCG neurons obtained from newborn mice transgenic with BMCC1 in primary culture. Thus, BMCC1 is a new member of prognostic factors for NBL and may play an important role in regulating differentiation, survival and aggressiveness of the tumor cells.
Collapse
Affiliation(s)
- T Machida
- Division of Biochemistry, Chiba Cancer Center Research Institute, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Wira CR, Fahey JV, Sentman CL, Pioli PA, Shen L. Innate and adaptive immunity in female genital tract: cellular responses and interactions. Immunol Rev 2005; 206:306-35. [PMID: 16048557 DOI: 10.1111/j.0105-2896.2005.00287.x] [Citation(s) in RCA: 370] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The mucosal immune system in the female reproductive tract (FRT) has evolved to meet the unique requirements of dealing with sexually transmitted bacterial and viral pathogens, allogeneic spermatozoa, and the immunologically distinct fetus. Analysis of the FRT indicates that the key cells of the innate and adaptive immune systems are present and functionally responsive to antigens. Acting through Toll-like receptors in the Fallopian tubes, uterus, cervix, and in the vagina, epithelial cells, macrophages, natural killer cells, and neutrophils confer protection through the production of chemokines and cytokines, which recruit and activate immune cells, as well as bactericidal and virucidal agents, which confer protection at times when adaptive immunity is downregulated by sex hormones to meet the constraints of procreation. The overall goal of this paper is to define the innate immune system in the FRT and, where possible, to define the regulatory influences that occur during the menstrual cycle that contribute to protection from and susceptibility to potential pathogens. By understanding the nature of this protection and the ways in which innate and adaptive immunity interact, these studies provide the opportunity to contribute to the foundation of information essential for ensuring reproductive health.
Collapse
Affiliation(s)
- Charles R Wira
- Department of Physiology, Dartmouth Medical School, Lebanon, NH 03756, USA.
| | | | | | | | | |
Collapse
|
44
|
Amantea D, Russo R, Bagetta G, Corasaniti MT. From clinical evidence to molecular mechanisms underlying neuroprotection afforded by estrogens. Pharmacol Res 2005; 52:119-32. [PMID: 15967377 DOI: 10.1016/j.phrs.2005.03.002] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Accepted: 03/14/2005] [Indexed: 11/15/2022]
Abstract
Recent studies have highlighted that female sex hormones represent potential neuroprotective agents against damage produced by acute and chronic injuries in the adult brain. Clinical reports have documented the effectiveness of estrogens to attenuate symptoms associated with Parkinson's disease, and to reduce the risk of Alzheimer's disease and cerebrovascular stroke. This evidence is corroborated by numerous experimental studies documenting the protective role of female sex hormones both in vitro and in vivo. Accordingly, estrogens have been shown to promote survival and differentiation of several neuronal populations maintained in culture, and to reduce cell death associated with excitotoxicity, oxidative stress, serum deprivation or exposure to beta-amyloid. The neuroprotective effects of estrogens have been widely documented in animal models of neurological disorders, such as Alzheimer's and Parkinson's diseases, as well as cerebral ischemia. Although estrogens are known to exert several direct effects on neurones, the cellular and molecular mechanisms implicated in their protective actions on the brain are not completely understood. Thus, on the basis of clinical and experimental evidence, in this review, we discuss recent findings concerning the neuronal effects of estrogens that may contribute to their neuroprotective actions. Both estrogen receptor-dependent and -independent mechanisms will be described. These include modulation of cell death regulators, such as Bcl-2, Akt and calpain, as well as interaction with growth factors, such as BDNF, NGF, IGF-I and their receptors. The anti-inflammatory effects of estrogens will also be described, namely their ability to reduce brain levels of inflammatory mediators, cytokines and chemokines. Finally, a brief overview about receptor-independent mechanisms of neuroprotection will aim at describing the antioxidant effects of estrogens, as well as their ability to modulate neurotransmission.
Collapse
Affiliation(s)
- Diana Amantea
- Department of Pharmacobiology, University of Calabria, Via P. Bucci, Ed. Polifunzionale, Arcavacata di Rende (CS), Italy
| | | | | | | |
Collapse
|
45
|
Jones NC, Constantin D, Prior MJW, Morris PG, Marsden CA, Murphy S. The neuroprotective effect of progesterone after traumatic brain injury in male mice is independent of both the inflammatory response and growth factor expression. Eur J Neurosci 2005; 21:1547-54. [PMID: 15845082 DOI: 10.1111/j.1460-9568.2005.03995.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Previous studies suggest that progesterone may possess neuroprotective properties after traumatic insult but, with the exception of reduced formation of cerebral oedema, limited experimental evidence has been presented to support this claim. In the present study we focused on the effect of progesterone treatment on structural and functional deficits in an experimental model of traumatic brain injury. Female mice exhibited significantly (P = 0.0445) reduced lesion volumes compared with males after aseptic cryogenic cerebral injury (ACI), suggesting that female sex steroids provide protection against this injury. In male mice, progesterone treatment after injury (three intraperitoneal doses of 8 mg/kg) reduced lesion volume (P = 0.0429) and improved performance in a spatial cognitive task (Morris water maze; P = 0.0014). However, progesterone had no demonstrable effect on the formation of oedema as measured using T2-weighted magnetic resonance imaging, nor did it affect brain water content. The pro-inflammatory cytokines TNF-alpha and IL-1beta, and growth factors BDNF and G-CSF, were all strongly transcriptionally activated after ACI. However, progesterone administration did not affect expression of these genes. This study provides strong evidence that progesterone possesses neuroprotective properties in a mouse model of traumatic brain injury, but suggests that the steroid achieves this effect through mechanism(s) independent of the inflammatory response or growth factor up-regulation.
Collapse
Affiliation(s)
- Nigel C Jones
- Institute of Cell Signalling, University of Nottingham, Clifton Blvd., Nottingham NG7 2UH, UK
| | | | | | | | | | | |
Collapse
|
46
|
Ghisletti S, Meda C, Maggi A, Vegeto E. 17beta-estradiol inhibits inflammatory gene expression by controlling NF-kappaB intracellular localization. Mol Cell Biol 2005; 25:2957-68. [PMID: 15798185 PMCID: PMC1069609 DOI: 10.1128/mcb.25.8.2957-2968.2005] [Citation(s) in RCA: 336] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Estrogen is an immunoregulatory agent, in that hormone deprivation increases while 17beta-estradiol (E2) administration blocks the inflammatory response; however, the underlying mechanism is still unknown. The transcription factor p65/relA, a member of the nuclear factor kappaB (NF-kappaB) family, plays a major role in inflammation and drives the expression of proinflammatory mediators. Here we report a novel mechanism of action of E2 in inflammation. We observe that in macrophages E2 blocks lipopolysaccharide-induced DNA binding and transcriptional activity of p65 by preventing its nuclear translocation. This effect is selectively activated in macrophages to prevent p65 activation by inflammatory agents and extends to other members of the NF-kappaB family, including c-Rel and p50. We observe that E2 activates a rapid and persistent response that involves the activation of phosphatidylinositol 3-kinase, without requiring de novo protein synthesis or modifying Ikappa-Balpha degradation and mitogen-activated protein kinase activation. Using a time course experiment and the microtubule-disrupting agent nocodazole, we observe that the hormone inhibits p65 intracellular transport to the nucleus. This activity is selectively mediated by estrogen receptor alpha (ERalpha) and not ERbeta and is not shared by conventional anti-inflammatory drugs. These results unravel a novel and unique mechanism for E2 anti-inflammatory activity, which may be useful for identifying more selective ligands for the prevention of the inflammatory response.
Collapse
Affiliation(s)
- Serena Ghisletti
- Center of Excellence on Neurodegenerative Diseases, University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | | | | | | |
Collapse
|
47
|
Lu B, Jiang YJ, Choy PC. 17-Beta estradiol enhances prostaglandin E2 production in human U937-derived macrophages. Mol Cell Biochem 2005; 262:101-10. [PMID: 15532714 DOI: 10.1023/b:mcbi.0000038222.08915.84] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Prostaglandins (PGs) are potent eicosanoid lipid mediators that have been implicated in numerous homeostatic functions and inflammation. Estrogens have been shown to regulate the expression of genes in lipid metabolism in many cellular systems. In this study, the activation of macrophages and the modulation of PG release by estrogens were examined. The effects of 17-alpha and 17-beta estradiols, phytoestrogen Genistein and several selective estrogen receptor modulators on the release of PGE2 were investigated in human U937-derived macrophages. 17-Beta estradiol caused an enhancement of PGE2 production in a time- and dose-dependent manner. Treatment of macrophages with 17-beta estradiol elicited an increased arachidonic acid (AA) release and an up-regulation of both cyclooxygenesis-1 and cyclooxygenesis-2 enzymes at both the transcript and protein levels. In addition, immunostaining of nuclear estrogen receptor alpha and the observation of ICI182 780 blockade of PGE2 production indicated that 17-beta estradiol-induced PGE2 release was mainly through nuclear estrogen receptor alpha.
Collapse
Affiliation(s)
- Biao Lu
- Department of Biochemistry and Medical Genetics, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | |
Collapse
|
48
|
Abstract
In addition to their effects on sexual differentiation and reproduction, sex hormones appear to influence the immune system. This results in a sexual dimorphism in the immune response in humans: for instance, females produce more vigorous cellular and more vigorous humoral immune reactions, are more resistant to certain infections, and suffer a higher incidence of autoimmune diseases. Disease expression is also affected by the reproductive status of the female. As sex steroids--estrogens, progesterone and testosterone--differ between gender and within different reproductive stages, a lot of research has focussed on the effects of sex hormones on immune responses. Although there is also a vast literature on the effects of sex hormones on immune responses in animals, in this review we will focus on the most intriguing effects and mechanisms by which sex hormones affect different components of the immune system in humans.
Collapse
Affiliation(s)
- Annechien Bouman
- Department of Obstetrics and Gynaecology, University Medical Centre Groningen, The Netherlands
| | | | | |
Collapse
|
49
|
Ramachandran S, Song MQ, Lowe E, Dominguez CE, Parthasarathy S, Murphy AA. RU486 inhibits expression of lysophosphatidic acid induced glycodelin. Am J Obstet Gynecol 2005; 192:1285-93; discussion 1293-4. [PMID: 15846221 DOI: 10.1016/j.ajog.2004.12.084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
OBJECTIVE This study was undertaken to provide evidence for the mode of action of RU486 on glycodelin produced in K562 cells. To show that histiocytes may be a source of glycodelin in leiomyoma. STUDY DESIGN With the use of K562, a leukemia cell line, the effect of lysophosphatidic acid (LPA), RU486, antioxidants, and ZK112,993 on glycodelin protein and gene expression was studied. Immunocytochemistry for glycodelin and HAM-56 (macrophage) was performed on leiomyoma and myometrium. RESULTS Incubation of K562 cells with LPA, progesterone, ZK112,933 and RU486 significantly induced the expression of glycodelin protein and messenger RNA. The addition of RU486 to LPA activated cells markedly reduced expression of glycodelin. Addition of ZK112,993, an antiprogestin without antioxidant properties, to LPA activated cells did not reduce glycodelin. Histiocytes in leiomyoma and myometrium co-localize with glycodelin. CONCLUSION RU486, partly acting as an antioxidant, markedly reduces LPA stimulated glycodelin production. Histiocytes in leiomyoma and myometrium immunostain for glycodelin and suggests a source for glycodelin in leiomyoma.
Collapse
Affiliation(s)
- Sumathi Ramachandran
- Emory University School of Medicine, Department of Gynecology and Obstetrics, Atlanta, GA, USA
| | | | | | | | | | | |
Collapse
|
50
|
García-Arencibia M, Dávila N, Campión J, Carmen Carranza M, Calle C. Identification of two functional estrogen response elements complexed with AP-1-like sites in the human insulin receptor gene promoter. J Steroid Biochem Mol Biol 2005; 94:1-14. [PMID: 15862945 DOI: 10.1016/j.jsbmb.2004.12.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This study was designed to explore the possible existence and location of estrogen response elements (EREs) in the human insulin receptor (hIR) gene promoter. Transfections of U-937 cells with the reported plasmids phIR(-1819)-GL2, phIR(-1473)-GL2, and phIR(-876)-GL2, that contain the -1819 to -271 bp fragment of the hIR promoter (wild-type promoter) and progressive 5' deletions of this promoter, revealed that while the activity of the wild-type promoter, was repressed 36% by treatment with 17beta-estradiol (E(2)), the activities of progressive 5' deletions of this promoter were reduced by 26% and by 0%, by this hormone. This suggests that E(2) needs the wild-type promoter for full transcriptional repression of this gene and it also suggests the presence of putative EREs in the region between -1819/-877 bp of this promoter. To identify these EREs we performed a computer search, using the SEQFIND programme developed in our laboratory, by homology with the consensus vit-ERE (5'GGTCAnnnTGACC3') of the Xenopus vitellogenin A(2) gene promoter. The results of our search indicated no sequence identical to this consensus ERE, and neither was any sequence found to show 9 or 8 of the 10 bases of this consensus in this promoter. Nevertheless, a putative hIR ERE1 (5'AGTGAaacTGGCC3') showing 7 bases of the consensus vit-ERE, and 10 bases of the optimal binding sequence ERE (5'CA/GGGTCAnnnTGACCT/CG3'), was identified between -1430/-1418bp of the hIR promoter. An AP-1-like site was covering the 3' half-element of this ERE; another AP-1-like site was overlapping the first AP-1-like site, and finally a third AP-1-like site was located beside to the 5' half-element. In addition, another putative hIR ERE2 (5'GCTCCtagCAAAC3') showing 5 bases of the consensus vit-ERE, and 9 bases of the optimal binding sequence ERE, was located upstream of the hIR promoter, between -1567/-1555 bp. An AP-1-like site was located downstream of the 3' half-element of this ERE, and another AP-1-like site was beside the 5' half-element. EMSA analysis using nuclear extracts of E(2)-treated cells and natural sequences, including these putative EREs, indicated that ERbeta - the only isoform expressed in U-937 cells - specifically recognized both EREs because ERbeta-DNA complexes were efficiently competed by the corresponding unlabelled probe and supershifted by the anti-human ERbeta (L-20) antibody. These data provide the first identification of EREs complexed with AP-1-like sites in the hIR promoter, which account for the transcriptional repression of the hIR gene mediated by ERbeta in U-937 cells.
Collapse
Affiliation(s)
- Moisés García-Arencibia
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University, 28040 Madrid, Spain
| | | | | | | | | |
Collapse
|