1
|
van Eeghen SA, Nokoff NJ, Vosters TG, Oosterom-Eijmael MJP, Cherney DZI, van Valkengoed IGM, Choi YJ, Pyle L, Bjornstad P, den Heijer M, van Raalte DH. Unraveling Sex Differences in Kidney Health and CKD: A Review of the Effect of Sex Hormones. Clin J Am Soc Nephrol 2024:01277230-990000000-00516. [PMID: 39671256 DOI: 10.2215/cjn.0000000642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
Sexual dimorphism plays an important role in the pathogenesis and progression of CKD. Men with CKD often exhibit faster kidney function decline, leading to higher rates of kidney failure and mortality compared with women. Studies suggest that sex hormones may influence this apparent dimorphism, although the mechanisms underlying these influences remain poorly understood. In this review, we first summarize recent findings on sex differences in the prevalence and progression of CKD. Subsequently, we will focus on ( 1 ) the role of sex hormones in these sex differences, ( 2 ) kidney structural and hemodynamic differences between men and women, ( 3 ) the influence of sex hormones on pathophysiological processes leading to kidney disease, including glomerular hyperfiltration and key pathways involved in kidney inflammation and fibrosis, and finally, focus on the consequences of the underrepresentation of women in clinical trials. Understanding these sex differences is critical for advancing precision medicine and improving outcomes for both men and women with CKD.
Collapse
Affiliation(s)
- Sarah A van Eeghen
- Department of Internal Medicine, Center of Expertise on Gender Dysphoria, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
| | - Natalie J Nokoff
- Section of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Taryn G Vosters
- Department of Public and Occupational Health, Amsterdam University Medical Centre, Universiteit van Amsterdam, Amsterdam, The Netherlands
| | - Maartina J P Oosterom-Eijmael
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
| | - David Z I Cherney
- Division of Nephrology, Department of Medicine, Toronto General Hospital, Toronto, Ontario, Canada
| | - Irene G M van Valkengoed
- Department of Public and Occupational Health, Amsterdam University Medical Centre, Universiteit van Amsterdam, Amsterdam, The Netherlands
| | - Ye Ji Choi
- Section of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, Colorado
| | - Laura Pyle
- Section of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, Colorado
| | - Petter Bjornstad
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
- Division of Endocrinology, Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington
| | - Martin den Heijer
- Department of Internal Medicine, Center of Expertise on Gender Dysphoria, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
| | - Daniël H van Raalte
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
- Department of Internal Medicine, Diabetes Center, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
- Amsterdam Cardiovascular sciences Research Institute, Amsterdam, the Netherlands
| |
Collapse
|
2
|
Husain A, Monga J, Narwal S, Singh G, Rashid M, Afzal O, Alatawi A, Almadani NM. Prodrug Rewards in Medicinal Chemistry: An Advance and Challenges Approach for Drug Designing. Chem Biodivers 2023; 20:e202301169. [PMID: 37833241 DOI: 10.1002/cbdv.202301169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/15/2023]
Abstract
This article emphasizes the importance of prodrugs and their diverse spectrum of effects in the field of developing novel drugs for a variety of biological applications. Prodrugs are chemicals that are supplied inactively, but then go through enzymatic and chemical transformation in vivo to release the active parent medication that can have the desired pharmacological effect. By adding an inactive chemical moiety, prodrugs are improved in a number of ways that contribute to their potency and durability. For the purpose of illustrating the usefulness of the prodrug approach, this review covers examples of prodrugs that have been made available or are now undergoing human trials. Additionally, it included lists of the most common functional groups, carrier linkers, and reactive chemicals that can be used to create prodrugs. The current study also provides a brief introduction, several chemical methods and modifications for creating prodrugs and mutual prodrugs, as well as an explanation of recent advancements and difficulties in the field of prodrug design. The primary chemical carriers employed in the creation of prodrugs, such as esters, amides, imides, NH-acidic carriers, amines, alcohols, carbonyl, carboxylic, and azo-linkages, are also discussed. This review also discusses glycosidic and triglyceride mutually activated prodrugs, which aim to deliver the drugs after bioconversion at the intended site of action. The article also discusses the extensive chemistry and wide variety of applications of recently approved prodrugs, such as antibacterial, anti-inflammatory, cardiovascular, antiplatelet, antihypertensive, atherosclerotic, antiviral, etc. In order to illustrate the prodrug and mutual drug concept's various applications and highlight its many triumphs in overcoming the formulation and delivery of problematic pharmaceuticals, this work represents a thorough guide that includes the synthetic moiety for the reader.
Collapse
Affiliation(s)
- Asif Husain
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110064, India
| | - Jyoti Monga
- Ch. Devi Lal College of Pharmacy, Jagadhri, 135003, Haryana, India
| | - Smita Narwal
- Global Research Institute of Pharmacy, Nachraun, Radaur, 135133, Haryana, India
| | - Gurvirender Singh
- Institute of Pharmaceutical Sciences, Kurukshetra University Kurukshetra-136119, Haryana, India
| | - Mohammad Rashid
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Dentistry and Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, 11942, Saudi Arabia
| | - Abdurahhman Alatawi
- Clinical Pharmacist, Pharmaceutical Care Department, King Fahad Specialized Hospital, Tabuk, 47717, Saudi Arabia
| | - Norah M Almadani
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk, 47914, Saudi Arabia
| |
Collapse
|
3
|
Exosomes Derived from Yak Follicular Fluid Increase 2-Hydroxyestradiol Secretion by Activating Autophagy in Cumulus Cells. Animals (Basel) 2022; 12:ani12223174. [PMID: 36428401 PMCID: PMC9686841 DOI: 10.3390/ani12223174] [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: 09/28/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022] Open
Abstract
Exosomes in the follicular fluid can carry and transfer regulatory molecules to recipient cells, thus influencing their biological functions. However, the specific effects of yak follicular fluid exosomes on 2-hydroxyestrodiol (2-OHE2) secretion remain unknown. Here, we investigated whether yak follicular fluid exosomes can increase 2-OHE2 secretion through the activation of autophagy in cumulus cells (YCCs). In vitro cultured YCCs were treated with yak follicular fluid exosomes for 6, 12, and 24 h. The effects of yak follicular fluid exosomes on autophagy and 2-OHE2 secretion were evaluated through real-time quantitative fluorescence PCR (RT-qPCR), Western blotting (WB), transfected with RFP-GFP-LC3, immunohistochemistry, and ELISA. To further investigate whether 2-OHE2 secretion was related to autophagy, YCCs were administered with yak follicular fluid exosomes, 3-methyladenine (3-MA), and rapamycin (RAPA). The results revealed that treatment with yak follicular fluid exosomes activated autophagy in YCCs and increased 2-OHE2 secretion. Conversely, the inhibition of autophagy with 3-MA blocked these effects, suggesting that autophagy has an important role in 2-OHE2 secretion in YCCs. Treatment of YCCs with rapamycin showed similar results with yak follicular fluid exosomes as there was an increase in 2-OHE2 secretion due to the activation of autophagy in the treated cumulus cells. Our results demonstrate that autophagy is enhanced by yak follicular fluid exosomes, and this is associated with an increase in 2-OHE2 secretion in YCCs.
Collapse
|
4
|
2-Methoxyestradiol Attenuates Angiotensin II-Induced Hypertension, Cardiovascular Remodeling, and Renal Injury. J Cardiovasc Pharmacol 2020; 73:165-177. [PMID: 30839510 DOI: 10.1097/fjc.0000000000000649] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Estradiol may antagonize the adverse cardiovascular effects of angiotensin II (Ang II). We investigated the effects of 2-methoxyestradiol (2-ME), a nonestrogenic estradiol metabolite, on Ang II-induced cardiovascular and renal injury in male rats. First, we determined the effects of 2-ME on Ang II-induced acute changes in blood pressure, renal hemodynamics, and excretory function. Next, we investigated the effects of 2-ME and 2-hydroxyestardiol (2-HE) on hypertension and cardiovascular and renal injury induced by chronic infusion of Ang II. Furthermore, the effects of 2-ME on blood pressure and cardiovascular remodeling in the constricted aorta (CA) rat model and on isoproterenol-induced (ISO) cardiac hypertrophy and fibrosis were examined. 2-ME had no effects on Ang II-induced acute changes in blood pressure, renal hemodynamics, or glomerular filtration rate. Both 2-ME and 2-HE reduced hypertension, cardiac hypertrophy, proteinuria, and mesangial expansion induced by chronic Ang II infusions. In CA rats, 2-ME attenuated cardiac hypertrophy and fibrosis and reduced elevated blood pressure above the constriction. Notably, 2-ME reduced both pressure-dependent (above constriction) and pressure-independent (below constriction) vascular remodeling. 2-ME had no effects on ISO-induced renin release yet reduced ISO-induced cardiac hypertrophy and fibrosis. This study shows that 2-ME protects against cardiovascular and renal injury due to chronic activation of the renin-angiotensin system. This study reports for the first time that in vivo 2-ME reduces trophic (pressure-independent) effects of Ang II and related cardiac and vascular remodeling.
Collapse
|
5
|
Neugarten J, Golestaneh L. Influence of Sex on the Progression of Chronic Kidney Disease. Mayo Clin Proc 2019; 94:1339-1356. [PMID: 31272577 DOI: 10.1016/j.mayocp.2018.12.024] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/21/2018] [Accepted: 12/17/2018] [Indexed: 12/14/2022]
Abstract
The role that sex plays in the development and progression of chronic kidney disease remains a subject of controversy. The lack of clarity in this important area reflects complex interactions between biological factors and cultural and socioeconomic influences that impact the relationship between sex and renal disease. Certainly, additional observational studies are indicated; however, innovative approaches are required to isolate biological processes from cultural influences. Despite these limitations, available data suggest that the progression of renal disease is slower in women than in men and that this sexual dimorphism is primarily due to direct actions of sex hormones on cellular metabolism. The extent to which differences in lifestyle factors between the sexes influence sexual dimorphism in the progression of chronic kidney disease remains to be elucidated.
Collapse
Affiliation(s)
- Joel Neugarten
- Albert Einstein College of Medicine, Renal Division, Montefiore Medical Center, Bronx, NY.
| | - Ladan Golestaneh
- Albert Einstein College of Medicine, Renal Division, Montefiore Medical Center, Bronx, NY
| |
Collapse
|
6
|
Vorland CJ, Lachcik PJ, Swallow EA, Metzger CE, Allen MR, Chen NX, Moe SM, Hill Gallant KM. Effect of ovariectomy on the progression of chronic kidney disease-mineral bone disorder (CKD-MBD) in female Cy/+ rats. Sci Rep 2019; 9:7936. [PMID: 31138895 PMCID: PMC6538713 DOI: 10.1038/s41598-019-44415-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/16/2019] [Indexed: 01/08/2023] Open
Abstract
Male Cy/+ rats have shown a relatively consistent pattern of progressive kidney disease development that displays multiple key features of late stage chronic kidney disease-mineral bone disorder (CKD-MBD), specifically the development of cortical bone porosity. However, progression of disease in female Cy/+ rats, assessed in limited studies, is more heterogeneous and to date has failed to show development of the CKD-MBD phenotype, thus limiting their use as a practical model of progressive CKD-MBD. Animal and human studies suggest that estrogen may be protective against kidney disease in addition to its established protective effect on bone. Therefore, in this study, we aimed to determine the effect of ovariectomy (OVX) on the biochemical and skeletal manifestations of CKD-MBD in Cy/+ female rats. We hypothesized that OVX would accelerate development of the biochemical and skeletal features of CKD-MBD in female Cy/+ rats, similar to those seen in male Cy/+ rats. Female Cy/+ rats underwent OVX (n = 8) or Sham (n = 8) surgery at 15 weeks of age. Blood was collected every 5 weeks post-surgery until 35 weeks of age, when the rats underwent a 4-day metabolic balance, and the tibia and final blood were collected at the time of sacrifice. OVX produced the expected changes in trabecular and cortical parameters consistent with post-menopausal disease, and negative phosphorus balance compared with Sham. However, indicators of CKD-MBD were similar between OVX and Sham (similar kidney weight, plasma blood urea nitrogen, creatinine, creatinine clearance, phosphorus, calcium, parathyroid hormone, and no cortical porosity). Contrary to our hypothesis, OVX did not produce evidence of development of the CKD-MBD phenotype in female Cy/+ rats.
Collapse
Affiliation(s)
- Colby J Vorland
- Department of Nutrition Science, Purdue University, West Lafayette, IN, 47907, USA
| | - Pamela J Lachcik
- Department of Nutrition Science, Purdue University, West Lafayette, IN, 47907, USA
| | - Elizabeth A Swallow
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
| | - Corinne E Metzger
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
| | - Matthew R Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
- Roudebush Veterans Affairs Medical Center, Indianapolis, IN, 46202, USA
| | - Neal X Chen
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
| | - Sharon M Moe
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, 46202, IN, USA
- Roudebush Veterans Affairs Medical Center, Indianapolis, IN, 46202, USA
| | - Kathleen M Hill Gallant
- Department of Nutrition Science, Purdue University, West Lafayette, IN, 47907, USA.
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, 46202, IN, USA.
| |
Collapse
|
7
|
Borges Canha M, Portela-Cidade JP, Conceição G, Sousa-Mendes C, Leite S, Fontoura D, Moreira-Gonçalves D, Falcão-Pires I, Lourenço A, Leite-Moreira A, Pimentel-Nunes P. Characterization of liver changes in ZSF1 rats, an animal model of metabolic syndrome. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2018; 109:491-497. [PMID: 28593786 DOI: 10.17235/reed.2017.4575/2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND The non-alcoholic fatty liver disease is the hepatic counterpart of the metabolic syndrome. ZSF1 rats are a metabolic syndrome animal model in which liver changes have not been described yet. AIM The characterization of liver histological and innate immunity changes in ZSF1 rats. METHODS Five groups of rats were included (n = 7 each group): healthy Wistar-Kyoto control rats (Ctrl), hypertensive ZSF1 lean (Ln), ZSF1 obese rats with a normal diet (Ob), ZSF1 obese rates with a high-fat diet (Ob-HFD), and ZSF1 obese rats with low-intensity exercise training (Ob-Ex). The animals were sacrificed at 20 weeks of age, their livers were collected for: a) measurements of the area of steatosis, fibrosis and inflammation (histomorphological analysis); and b) innate immunity (toll-like receptor [TLR] 2, TLR4, peroxisome proliferator-activated receptor γ [PPARγ], toll interacting protein [TOLLIP]) and inflammatory marker (tumor necrosis factor-alpha [TNFα], interleukin 1 [IL-1]) expression analysis by real-time PCR. RESULTS Ob, Ob-HFD and Ob-Ex were significantly heavier than Ln and Ctrl animals. Ob, Ob-HFD and Ob-Ex animals had impaired glucose tolerance and insulin resistance. ZSF1 Ob, Ob-HFD and Ob-Ex presented a higher degree of steatosis (3,5x; p < 0.05) than Ctrl or ZSF1 Ln rats. Steatohepatitis and fibrosis were not observed in any of the groups. No differences in expression were observed between Ctrl, Ln and Ob animals (except for the significantly higher expression of TOLLIP observed in the Ob vs Ln comparison). Ob-HFD and Ob-Ex rats showed increased expression of PPARγ and TOLLIP as compared to other groups. However, both groups also showed increased expression of TLR2 and TLR4. Nevertheless, this did not translate into a differential expression of TNFα or IL-1 in any of the groups. CONCLUSION The ZSF1 model is associated with liver steatosis but not with steatohepatitis or a significantly increased expression of innate immunity or inflammation markers.
Collapse
Affiliation(s)
- Marta Borges Canha
- Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Portugal
| | | | - Glória Conceição
- Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto
| | | | - Sara Leite
- Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto
| | - Dulce Fontoura
- Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto
| | | | - Inês Falcão-Pires
- Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto
| | - André Lourenço
- Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto
| | | | | |
Collapse
|
8
|
Mirfazeli ES, Marashi SA, Kalantari S. In silico prediction of specific pathways that regulate mesangial cell proliferation in IgA nephropathy. Med Hypotheses 2016; 97:38-45. [PMID: 27876127 DOI: 10.1016/j.mehy.2016.10.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 09/28/2016] [Accepted: 10/19/2016] [Indexed: 11/30/2022]
Abstract
IgA nephropathy is one of the most common forms of primary glomerulonephritis worldwide leading to end-stage renal disease. Proliferation of mesangial cells, i.e., the multifunctional cells located in the intracapillary region of glomeruli, after IgA- dominant immune deposition is the major histologic feature in IgA nephropathy. In spite of several studies on molecular basis of proliferation in these cells, specific pathways responsible for regulation of proliferation are still to be discovered. In this study, we predicted a specific signaling pathway started from transferrin receptor (TFRC), a specific IgA1 receptor on mesangial cells, toward a set of proliferation-related proteins. The final constructed subnetwork was presented after filtration and evaluation. The results suggest that estrogen receptor (ESR1) as a hub protein in the significant subnetwork has an important role in the mesangial cell proliferation and is a potential target for IgA nephropathy therapy. In conclusion, this study suggests a novel hypothesis for the mechanism of pathogenesis in IgA nephropathy and is a reasonable start point for the future experimental studies on mesangial proliferation process in this disease.
Collapse
Affiliation(s)
| | - Sayed-Amir Marashi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Shiva Kalantari
- Chronic Kidney Disease Research Center (CKDRC), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
An integrated lipidomics and metabolomics reveal nephroprotective effect and biochemical mechanism of Rheum officinale in chronic renal failure. Sci Rep 2016; 6:22151. [PMID: 26903149 PMCID: PMC4763304 DOI: 10.1038/srep22151] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/08/2016] [Indexed: 02/08/2023] Open
Abstract
Chronic renal failure (CRF) is a major public health problem worldwide. Earlier studies have revealed salutary effects of rhubarb extracts in CRF. In this study, we employed lipidomic and metabolomic approaches to identify the plasma biomarkers and to determine the effect of treatment with petroleum ether, ethyl acetate and n-butanol extracts of rhubarb in a rat model of CRF with adenine-induced chronic tubulointerstitial nephropathy. In addition, clinical biochemistry, histological evaluation and pro-fibrotic protein expression were analyzed. Significant changes were found between the CRF and control groups representing characteristic phenotypes of rats with CRF. Treatment with the three rhubarb extracts improved renal injury and dysfunction, either fully or partially reversed the plasma metabolites abnormalities and attenuated upregulation of pro-fibrotic proteins including TGF-β1, α-SMA, PAI-1, CTGF, FN and collagen-1. The nephroprotective effect of ethyl acetate extract was better than other extracts. The differential metabolites were closely associated with glycerophospholipid, fatty acid and amino acid metabolisms. The results revealed a strong link between renal tubulointerstitial fibrosis and glycerophospholipid metabolism and L-carnitine metabolism in the development of CRF. Amelioration of CRF with the three rhubarb extracts was associated with the delayed development and/or reversal the disorders in key metabolites associated with adenine-induced CRF.
Collapse
|
10
|
Lahm T, Tuder RM, Petrache I. Progress in solving the sex hormone paradox in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2014; 307:L7-26. [PMID: 24816487 DOI: 10.1152/ajplung.00337.2013] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating and progressive disease with marked morbidity and mortality. Even though being female represents one of the most powerful risk factors for PAH, multiple questions about the underlying mechanisms remain, and two "estrogen paradoxes" in PAH exist. First, it is puzzling why estrogens have been found to be protective in various animal models of PAH, whereas PAH registries uniformly demonstrate a female susceptibility to the disease. Second, despite the pronounced tendency for the disease to develop in women, female PAH patients exhibit better survival than men. Recent mechanistic studies in classical and in novel animal models of PAH, as well as recent studies in PAH patients, have significantly advanced the field. In particular, it is now accepted that estrogen metabolism and receptor signaling, as well as estrogen interactions with key pathways in PAH development, appear to be potent disease modifiers. A better understanding of these interactions may lead to novel PAH therapies. It is the purpose of this review to 1) review sex hormone synthesis, metabolism, and receptor physiology; 2) assess the context in which sex hormones affect PAH pathogenesis; 3) provide a potential explanation for the observed estrogen paradoxes and gender differences in PAH; and 4) identify knowledge gaps and future research opportunities. Because the majority of published studies investigated 17β-estradiol and/or its metabolites, this review will primarily focus on pulmonary vascular and right ventricular effects of estrogens. Data for other sex hormones will be discussed very briefly.
Collapse
Affiliation(s)
- Tim Lahm
- Division of Pulmonary, Allergy, Critical Care, Occupational and Sleep Medicine, and Richard L. Roudebush VA Medical Center; Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Rubin M Tuder
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, School of Medicine, Denver, Colorado
| | - Irina Petrache
- Division of Pulmonary, Allergy, Critical Care, Occupational and Sleep Medicine, and Richard L. Roudebush VA Medical Center; Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| |
Collapse
|
11
|
Hall YN, Choi AI, Xu P, Smith NL, Boyko EJ. Predictors of end-stage renal disease in the urban poor. J Health Care Poor Underserved 2013; 24:1686-700. [PMID: 24185164 PMCID: PMC4504426 DOI: 10.1353/hpu.2013.0189] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We sought to examine the influence of social and clinical factors on risk of progression of chronic kidney disease (CKD) to end-stage renal disease (ESRD) in the urban poor. We studied 15,353 individuals with moderate-to-advanced CKD who received ambulatory care within a large public health system during 1996-2005. The primary outcome was progression to ESRD. Overall, 559 cases of ESRD occurred over a median follow-up of 2.8 years. Among traditional predictors of ESRD, younger age, male sex, non-White race/ethnicity, public health insurance coverage, diabetes, lower kidney function, higher proteinuria, lower hemoglobin level, and lower serum albumin concentration were significantly associated with a higher adjusted ESRD risk (p<.001 for all). There was no significant association between HIV/AIDS (p=.07), viral hepatitis (p=.11), or non-English language (p=.27) and ESRD risk. Our results highlight the importance of addressing traditional risk factors for progressive CKD to reduce the disproportionate burden of ESRD among disadvantaged populations.
Collapse
|
12
|
Austin ED, Lahm T, West J, Tofovic SP, Johansen AK, MacLean MR, Alzoubi A, Oka M. Gender, sex hormones and pulmonary hypertension. Pulm Circ 2013; 3:294-314. [PMID: 24015330 PMCID: PMC3757824 DOI: 10.4103/2045-8932.114756] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Most subtypes of pulmonary arterial hypertension (PAH) are characterized by a greater susceptibility to disease among females, although females with PAH appear to live longer after diagnosis. While this "estrogen paradoxȍ of enhanced female survival despite increased female susceptibility remains a mystery, recent progress has begun to shed light upon the interplay of sex hormones, the pathogenesis of pulmonary hypertension, and the right ventricular response to stress. For example, emerging data in humans and experimental models suggest that estrogens or differential sex hormone metabolism may modify disease risk among susceptible subjects, and that estrogens may interact with additional local factors such as serotonin to enhance the potentially damaging chronic effects of estrogens on the pulmonary vasculature. Regardless, it remains unclear why not all estrogenic compounds behave equally, nor why estrogens appear to be protective in certain settings but detrimental in others. The contribution of androgens and other compounds, such as dehydroepiandrosterone, to pathogenesis and possibly treatment must be considered as well. In this review, we will discuss the recent understandings on how estrogens, estrogen metabolism, dehydroepiandrosterone, and additional susceptibility factors may all contribute to the pathogenesis or potentially to the treatment of pulmonary hypertension, by evaluating current human, cell-based, and experimental model data.
Collapse
Affiliation(s)
- Eric D. Austin
- Department of Pediatrics, Division of Allergy, Immunology, and Pulmonary Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Tim Lahm
- Division of Pulmonary, Allergy, Critical Care, Occupational, and Sleep Medicine and Richard L. Roudebush Veterans Affairs Medical Center, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - James West
- Department of Medicine, Division of Allergy, Immunology, and Pulmonary Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Stevan P. Tofovic
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anne Katrine Johansen
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, USA
| | - Margaret R. MacLean
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, USA
| | - Abdallah Alzoubi
- Department of Medicine and Pharmacology and Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
| | - Masahiko Oka
- Department of Medicine and Pharmacology and Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
13
|
The influence of gender and sexual hormones on incidence and outcome of chronic kidney disease. Pediatr Nephrol 2012; 27:1213-9. [PMID: 21766172 DOI: 10.1007/s00467-011-1963-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 06/19/2011] [Accepted: 06/19/2011] [Indexed: 01/03/2023]
Abstract
It has long been known that the female sex is associated with a better clinical outcome in chronic renal diseases. Although many experimental, clinical, and epidemiological studies in adults have attempted to explain the difference in disease progression between females and males, a definitive understanding of the underlying mechanisms is still lacking. Hormone-modulating therapies are being increasingly used for various indications (such as post-menopausal hormone replacement, estrogen- or androgen-receptor antagonists for cancer therapy). Therefore, a deeper knowledge of the interaction between sexual hormones and progression of kidney disease is important, as hormone-modulating therapy for non-renal indication may influence both kidney structure and function. In addition, specific modulation of the sexual hormone system, such as the use of selective estrogen receptor modulators, may represent a therapeutic option for patients with renal diseases. Although conclusive data on this topic in the pediatric population are still lacking, the aim of this review is to familiarize pediatric nephrologists with gender-specific differences in renal physiology, pathophysiology, and the progression of kidney diseases. Experimental models that analyze the effects of sexual hormones on renal structure and function are discussed. It is hoped that this review will stimulate researchers to focus on pediatric studies that will provide a deeper insight into the interaction of gender hormones and the kidney both before and during puberty.
Collapse
|
14
|
Anderson S, Oyama TT, Lindsley JN, Schutzer WE, Beard DR, Gattone VH, Komers R. 2-Hydroxyestradiol slows progression of experimental polycystic kidney disease. Am J Physiol Renal Physiol 2011; 302:F636-45. [PMID: 22160773 DOI: 10.1152/ajprenal.00265.2011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Male gender is a risk factor for progression of polycystic kidney disease (PKD). 17β-Estradiol (E2) protects experimentally, but clinical use is limited by adverse effects. Novel E2 metabolites provide many benefits of E2 without stimulating the estrogen receptor, and thus may be safer. We hypothesized that E2 metabolites are protective in a model of PKD. Studies were performed in male control Han:SPRD rats, and in cystic males treated with orchiectomy, 2-methoxyestradiol, 2-hydroxyestradiol (2-OHE), or vehicle, from age 3 to 12 wk. Cystic rats exhibited renal functional impairment (∼50% decrease in glomerular filtration and renal plasma flow rates, P < 0.05) and substantial cyst development (20.5 ± 2.0% of cortex area). 2-OHE was the most effective in limiting cysts (6.0 ± 0.7% of cortex area, P < 0.05 vs. vehicle-treated cystic rats) and preserving function, in association with suppression of proliferation, apoptosis, and angiogenesis markers. Downregulation of p21 expression and increased expression of Akt, the mammalian target of rapamycin (mTOR), and some of its downstream effectors were significantly reversed by 2-OHE. Thus, 2-OHE limits disease progression in a cystic rodent model. Mechanisms include reduced renal cell proliferation, apoptosis, and angiogenesis. These effects may be mediated, at least in part, by preservation of p21 and suppression of Akt and mTOR. Estradiol metabolites may represent a novel, safe intervention to slow progression of PKD.
Collapse
Affiliation(s)
- Sharon Anderson
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Kummer S, Jeruschke S, Wegerich LV, Peters A, Lehmann P, Seibt A, Mueller F, Koleganova N, Halbenz E, Schmitt CP, Bettendorf M, Mayatepek E, Gross-Weissmann ML, Oh J. Estrogen receptor alpha expression in podocytes mediates protection against apoptosis in-vitro and in-vivo. PLoS One 2011; 6:e27457. [PMID: 22096576 PMCID: PMC3214053 DOI: 10.1371/journal.pone.0027457] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 10/17/2011] [Indexed: 11/18/2022] Open
Abstract
CONTEXT/OBJECTIVE Epidemiological studies have demonstrated that women have a significantly better prognosis in chronic renal diseases compared to men. This suggests critical influences of gender hormones on glomerular structure and function. We examined potential direct protective effects of estradiol on podocytes. METHODS Expression of estrogen receptor alpha (ERα) was examined in podocytes in vitro and in vivo. Receptor localization was shown using Western blot of separated nuclear and cytoplasmatic protein fractions. Podocytes were treated with Puromycin aminonucleoside (PAN, apoptosis induction), estradiol, or both in combination. Apoptotic cells were detected with Hoechst nuclear staining and Annexin-FITC flow cytometry. To visualize mitochondrial membrane potential depolarization as an indicator for apoptosis, cells were stained with tetramethyl rhodamine methylester (TMRM). Estradiol-induced phosphorylation of ERK1/2 and p38 MAPK was examined by Western blot. Glomeruli of ERα knock-out mice and wild-type controls were analysed by histomorphometry and immunohistochemistry. RESULTS ERα was consistently expressed in human and murine podocytes. Estradiol stimulated ERα protein expression, reduced PAN-induced apoptosis in vitro by 26.5±24.6% or 56.6±5.9% (flow cytometry or Hoechst-staining, respectively; both p<0.05), and restored PAN-induced mitochondrial membrane potential depolarization. Estradiol enhanced ERK1/2 phosphorylation. In ERα knockout mice, podocyte number was reduced compared to controls (female/male: 80/86 vs. 132/135 podocytes per glomerulus, p<0.05). Podocyte volume was enhanced in ERα knockout mice (female/male: 429/371 µm(3) vs. 264/223 µm(3) in controls, p<0.05). Tgfβ1 and collagen type IV expression were increased in knockout mice, indicating glomerular damage. CONCLUSIONS Podocytes express ERα, whose activation leads to a significant protection against experimentally induced apoptosis. Possible underlying mechanisms include stabilization of mitochondrial membrane potential and activation of MAPK signalling. Characteristic morphological changes indicating glomerulopathy in ERα knock-out mice support the in vivo relevance of the ERα for podocyte viability and function. Thus, our findings provide a novel model for the protective influence of female gender on chronic glomerular diseases.
Collapse
Affiliation(s)
- Sebastian Kummer
- Department of General Pediatrics and Neonatology, University Children's Hospital Duesseldorf, Duesseldorf, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Raloxifene, a selective estrogen receptor modulator, is renoprotective: a post-hoc analysis. Kidney Int 2010; 79:241-9. [PMID: 20927038 DOI: 10.1038/ki.2010.378] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Estrogens have a protective effect on kidney fibrosis in several animal models. Here, we tested the effect of raloxifene, an estrogen receptor modulator, on the change in serum creatinine or estimated glomerular filtration rate (eGFR) and incident kidney-related adverse events. We performed a post-hoc analysis of the multiple outcomes of raloxifene evaluation trial, a double-masked, placebo-controlled randomized clinical trial encompassing 7705 post-menopausal women (aged 31-80 years) with osteoporosis. Participants were randomized to either of two doses of raloxifene, 60 or 120 mg/day, or placebo. Serum creatinine was measured at a central laboratory at baseline and annually. Adverse events were assessed every 6 months and uniformly categorized. Compared with those in the placebo group, participants on raloxifene had a slower yearly rate of increase in creatinine (significant at the low dose) and a significantly slower yearly rate of decrease in eGFR for both doses over 3 years of follow-up. Raloxifene was associated with significantly fewer kidney-related adverse events compared with placebo. Thus, treatment with raloxifene was safe and renoprotective. Clinical trials of raloxifene in post-menopausal women with kidney disease designed to look at kidney outcomes are needed to confirm these findings.
Collapse
|
17
|
Schmidt M, Hartung R, Capellino S, Cutolo M, Pfeifer-Leeg A, Straub RH. Estrone/17β-estradiol conversion to, and tumor necrosis factor inhibition by, estrogen metabolites in synovial cells of patients with rheumatoid arthritis and patients with osteoarthritis. ACTA ACUST UNITED AC 2009; 60:2913-22. [DOI: 10.1002/art.24859] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
18
|
Sandros MG, Sarraf CB, Tabrizian M. Prodrugs in cardiovascular therapy. Molecules 2008; 13:1156-78. [PMID: 18560335 PMCID: PMC6245309 DOI: 10.3390/molecules13051156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Revised: 05/14/2008] [Accepted: 05/14/2008] [Indexed: 11/27/2022] Open
Abstract
Prodrugs are biologically inactive derivatives of an active drug intended to solve certain problems of the parent drug such as toxicity, instability, minimal solubility and non-targeting capabilities. The majority of drugs for cardiovascular diseases undergo first-pass metabolism, resulting in drug inactivation and generation of toxic metabolites, which makes them appealing targets for prodrug design. Since prodrugs undergo a chemical reaction to form the parent drug once inside the body, this makes them very effective in controlling the release of a variety of compounds to the targeted site. This review will provide the reader with an insight on the latest developments of prodrugs that are available for treating a variety of cardiovascular diseases. In addition, we will focus on several drug delivery methodologies that have merged with the prodrug approach to provide enhanced target specificity and controlled drug release with minimal side effects.
Collapse
Affiliation(s)
- Marinella G. Sandros
- Department of Biomedical Engineering, McGill University, 3775 University Street, Montreal, QC, Canada H3A2B4
- Center for Biorecognition and Biosensors, McGill Institute for Advanced Materials, 3775 University Street, Montreal, QC, Canada H3A2B4
| | - Chady B. Sarraf
- Department of Medical Education, Seton Hall University, 400 South Orange Avenue, South Orange, NJ 07079, USA
- St. Michael’s Medical Center, 111 Central Avenue, Newark, NJ 070102, USA
| | - Maryam Tabrizian
- Department of Biomedical Engineering, McGill University, 3775 University Street, Montreal, QC, Canada H3A2B4
- Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC, Canada, H3A 2B2
- Center for Biorecognition and Biosensors, McGill Institute for Advanced Materials, 3775 University Street, Montreal, QC, Canada H3A2B4
- Author to whom correspondence should be addressed; E-Mail:
| |
Collapse
|
19
|
Tofovic SP, Zhang X, Zhu H, Jackson EK, Rafikova O, Petrusevska G. 2-Ethoxyestradiol is antimitogenic and attenuates monocrotaline-induced pulmonary hypertension and vascular remodeling. Vascul Pharmacol 2008; 48:174-83. [DOI: 10.1016/j.vph.2008.02.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Revised: 01/27/2008] [Accepted: 02/04/2008] [Indexed: 10/22/2022]
|
20
|
Hiong LC, Voon KL, Abdullah NA, Sattar MA, Rahman NA, Khan AH, Johns EJ. Effect of TGF-beta1 antisense oligodeoxynucleotide on renal function in chronic renal failure rats. Acta Pharmacol Sin 2008; 29:451-7. [PMID: 18358091 DOI: 10.1111/j.1745-7254.2008.00772.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
AIM The aim of the present study was to investigate the effectiveness of transforming growth factor (TGF)-beta1 antisense oligodeoxynucleotides (ODN) in ameliorating deteriorated kidney function in rats with puromycin-induced chronic renal failure (CRF). METHODS Saline, puromycin, puromycin+TGF-beta1 antisense ODN or puromycin+scrambled ODN were administered to unilaterally nephrectomized rats. Renal hemodynamic and excretory measurements were taken in the anaesthetized rats that had undergone surgical procedure. RESULTS It was observed that in the CRF rats, there was a marked reduction in the renal blood flow (RBF), glomerular filtration rate (GFR), severe proteinuria, and almost 6-fold increased fractional excretion of sodium (FE Na+) as compared to that in the control rats (all P<0.05). It was further observed that in the CRF rats, the treatment with TGF-beta1 antisense, but not scrambled ODN, markedly attenuated the reduction of RBF, GFR, and proteinuria and markedly prevented the increase of the FE Na+ (all P<0.05). In addition, the renal hypertrophy in the CRF group (P<0.05 vs non-renal failure control) was markedly attenuated after treatment with TGF-1 antisense ODN (P<0.05). Focal segmental glomerulosclerosis was evident only in the untreated and scrambled ODN-treated CRF groups. An interesting observation of this study was that in the CRF rats, although there was marked attenuating and preventive effects of the TGF-beta1 antisense ODN on the deteriorated renal functions, the antisense treatment did not cause any marked change in the renal expression of TGF-beta1 at the protein level. CONCLUSION Collectively, the data obtained suggests that TGF-beta1 antisense ODN possesses beneficial effects in puromycininduced chronic renal failure and that the deterioration in morphology and impaired renal function in this pathological state is in part dependent upon the action of TGF-beta1 within the kidney.
Collapse
Affiliation(s)
- Law Chung Hiong
- Department of Pharmacology, University of Malaya, Kuala Lumpur 50603, Malaysia
| | | | | | | | | | | | | |
Collapse
|
21
|
Issekutz AC, Sapru K. Modulation of adjuvant arthritis in the rat by 2-methoxyestradiol: an effect independent of an anti-angiogenic action. Int Immunopharmacol 2008; 8:708-16. [PMID: 18387513 DOI: 10.1016/j.intimp.2008.01.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Revised: 01/15/2008] [Accepted: 01/16/2008] [Indexed: 12/13/2022]
Abstract
Angiogenesis is a prominent feature in rheumatoid arthritis. 2-methoxyestradiol (2ME2) inhibits endothelial cell proliferation, and angiogenesis in vivo. We evaluated the effect of 2ME2 in rats with adjuvant arthritis (AA), an autoimmune T-cell-dependent polyarticular arthritis induced by immunization with Mycobacterium organisms. Rats were immunized with Mycobacterium butyricum and arthritis was assessed clinically, by radiolabeled blood neutrophil (PMNL) migration to joints and by histology. Treatment with 2ME2 (30 mg/kg/d or 100 mg/kg/d) from day 6 post-immunization inhibited arthritis severity on day 14 (vehicle clinical score=11.2; 2ME2 groups=7-8, p<0.05). When treatment was delayed until signs of clinical arthritis on day 10 post-immunization, 2ME2 treatment still inhibited arthritis severity. PMNL migration to the joints was significantly inhibited (by 35-40%; p<0.01) by early 2ME2 treatment (day 6-14). Treatment with 2ME2 inhibited PMNL migration to dermal inflammation induced by TNF-alpha but not by LPS or C5a. Joint histology revealed decrease in leukocyte infiltration and especially in cartilage damage. However, synovial vascularity was not affected by 2ME2 treatment. The marked splenomegaly, splenitis and lymphoid hyperplasia associated with AA were prevented by 2ME2 therapy. Furthermore, the ex vivo proliferative response to mycobacterial antigen (PPD) of lymphocytes from 2ME2-treated rats with AA was markedly diminished, although response to mitogens was unaffected. Thus 2ME2 has anti-arthritic properties with a disease-modifying action, separate from its anti-angiogenic properties. The selective inhibition of TNF-alpha-induced leukocyte recruitment, lymphoid hyperplasia and attenuated recall response to antigen suggests both immunomodulatory and anti-inflammatory actions of 2ME2.
Collapse
Affiliation(s)
- Andrew C Issekutz
- Department of Pediatrics Dalhousie University, Halifax, Nova Scotia, Canada.
| | | |
Collapse
|
22
|
Dubey RK, Imthurn B, Jackson EK. 2-Methoxyestradiol: a potential treatment for multiple proliferative disorders. Endocrinology 2007; 148:4125-7. [PMID: 17699679 DOI: 10.1210/en.2007-0514] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Raghvendra K Dubey
- Department of Obstetrics and Gynecology, Clinic for Reproductive Endocrinology, University Hospital Zurich, Frauenklinikstrasse 10, 8091 Zurich, Switzerland.
| | | | | |
Collapse
|
23
|
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
|
24
|
|
25
|
Zhang X, Jia Y, Jackson EK, Tofovic SP. 2-Methoxyestradiol and 2-Ethoxyestradiol Retard the Progression of Renal Disease in Aged, Obese, Diabetic ZSF1 Rats. J Cardiovasc Pharmacol 2007; 49:56-63. [PMID: 17261964 DOI: 10.1097/fjc.0b013e31802cb88e] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The metabolic syndrome is a main cause for cardiovascular disease and for the accelerating epidemic of chronic renal failure. Previous studies show that 2-hydroxyestradiol (2-HE), an estradiol metabolite with little estrogenic activity, decreases obesity and arterial blood pressure and attenuates the development of renal disease in young, obese, diabetic ZSF1 rats. In humans, however, diabetic renal disease is more frequent and severe in older patients. In vivo, 2-HE is readily converted to 2-methoxyestradiol (2-ME), an estradiol metabolite with no estrogenic activity. Accordingly, one purpose of this study was to determine whether 2-ME would provide benefit in aged rats with a very severe form of diabetic renal disease. Another objective was to determine whether synthetic analogs of estradiol metabolites might be beneficial in diabetic renal disease. To achieve these objectives we examined the effects of 2-ME and its analog 2-ethoxyestradiol (2-EE) in aged (35-week-old), obese ZSF1 rats. Animals were treated for 9 weeks with vehicle (PEG-400, 0.5 microL per hour), 2-ME or 2-EE (18 microg/kg per hour). Metabolic and renal function were measured at weeks 0, 3, 6, and 9, and renal hemodynamics and excretory function were assessed at week 9. Aged ZSF1 rats had elevated levels of glycosylated hemoglobin; increased renal cortical expression of proliferating cell nuclear antigen (PCNA), nuclear factor kappa B (NF-kappaB), and vascular endothelial growth factor (VEGF); glycosuria, hypertension; and proteinuria. 2-ME and 2-EE did not affect obesity or hypertension and had variable effects on glucose homeostasis, yet they attenuated proteinuria; increased renal blood flow and glomerular filtration; and reduced renal cortical expression of PCNA, NFkappaB, and VEGF. We conclude that 2ME and 2EE are strikingly renoprotective even in aged animals with severe diabetic renal disease. The present study warrants further investigation of 2-ME and analogs of estradiol metabolites for treatment of kidney disease associated with the metabolic syndrome.
Collapse
Affiliation(s)
- Xinchen Zhang
- Center for Clinical Pharmacolog, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | | | | | | |
Collapse
|
26
|
Mankhey RW, Wells CC, Bhatti F, Maric C. 17beta-Estradiol supplementation reduces tubulointerstitial fibrosis by increasing MMP activity in the diabetic kidney. Am J Physiol Regul Integr Comp Physiol 2006; 292:R769-77. [PMID: 16931652 DOI: 10.1152/ajpregu.00375.2006] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported that supplementation with 17beta-estradiol (E2) attenuates albuminuria, glomerulosclerosis, and tubulointerstitial fibrosis in diabetic nephropathy. The present study examined the mechanisms by which E2 regulates extracellular matrix (ECM) metabolism, a process that contributes to the development of glomerulosclerosis and tubulointerstitial fibrosis. The study was performed in female nondiabetic (ND), streptozotocin-induced diabetic (D), and diabetic with E2 supplementation (D+E2) Sprague-Dawley rats for 12 wk. Diabetes was associated with an increase in the renal expression of collagen alpha type IV [ND, 0.22 +/- 0.02; D, 0.99 +/- 0.09 relative optical density (ROD); P < 0.05] and fibronectin protein (ND, 0.36 +/- 0.08; D, 1.47 +/- 0.08 ROD; P < 0.05), as measured by Western blot analysis. E2 supplementation partially attenuated this increase in collagen alpha type IV (D+E2, 0.47 +/- 0.10 ROD) and fibronectin (D+E2, 0.71 +/- 0.16 ROD) protein expression associated with D. Diabetes was also associated with a decrease in the expression of matrix metalloproteinase (MMP) isoform MMP-2 (ND, 0.79 +/- 0.01; D, 0.62 +/- 0.06 ROD; P < 0.05) and MMP-9 protein (ND, 0.49 +/- 0.02; D, 0.33 +/- 0.03 ROD; P < 0.05). E2 supplementation restored MMP-2 and MMP-9 protein to levels similar or even greater than in the ND kidneys (MMP-2, 0.75 +/- 0.06; MMP-9, 0.73 +/- 0.01 ROD). The activities of MMP-2 (ND, 7.88 +/- 0.44; D, 5.60 +/- 0.54 ROD; P < 0.05) and MMP-9 (ND, 29.9 +/- 1.8; D, 12.9 +/- 2.3 ROD; P < 0.05), as measured by zymography, were also decreased with D. E2 supplementation restored MMP-2 and MMP-9 activity to levels similar to that in ND kidneys (MMP-2, 7.66 +/- 0.35; MMP-9, 21.4 +/- 2.9 ROD). We conclude that E2 supplementation is renoprotective by attenuating glomerulosclerosis and tubulointerstitial fibrosis by reducing ECM synthesis and increasing ECM degradation.
Collapse
Affiliation(s)
- Richard W Mankhey
- Department of Medicine, Georgetown Univ. Medical Center, 394 Bldg D, 4000 Reservoir Rd., NW, Washington, DC 20057, USA
| | | | | | | |
Collapse
|
27
|
Tofovic SP, Salah EM, Mady HH, Jackson EK, Melhem MF. Estradiol metabolites attenuate monocrotaline-induced pulmonary hypertension in rats. J Cardiovasc Pharmacol 2006; 46:430-7. [PMID: 16160593 DOI: 10.1097/01.fjc.0000175878.32920.17] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Pulmonary arterial hypertension (PH) is a deadly disease characterized by pulmonary arterial vasoconstriction and hypertension, pulmonary vasculature remodeling, and right ventricular hypertrophy. Our previous in vivo studies, performed in several models of cardiac, vascular, and/or renal injury, suggest that the metabolites of 17beta-estradiol may inhibit vascular and cardiac remodeling. The goal of this study was to determine whether 2-methoxyestradiol (2ME), major non-estrogenic estradiol metabolite, prevents the development and/or retards the progression of monocrotaline (MCT)-induced PH. First, a total of 27 male Sprague Dawley rats were injected with distillated water (Cont, n=6) or monocrotaline (MCT; 60 mg/kg, i.p.; n=21). Subsets of MCT animals (n=7 per group) received 2ME or its metabolic precursor 2-hydroxyestradiol (2HE; 10 microg/kg/h via osmotic minipumps) for 21 days. Next, an additional set (n=24) of control and MCT rats was monitored for 28 days, before right ventricular peak systolic pressure (RVPSP) was measured. Some pulmonary hypertensive animals (n=8) were treated with 2ME (10 microg/kg/h) beginning from day 14 after MCT administration. MCT caused pulmonary hypertension (ie, increased right ventricle/left ventricle+septum [RV/LV+S] ratio and wall thickness of small-sized pulmonary arteries, and elevated RVPSP) and produced high and late (days 22 to 27) mortality. Pulmonary hypertension was associated with strong proliferative response (PCNA staining) and marked inflammation (ED1+cells) in lungs. Both metabolites significantly attenuated the RV/LV+S ratio and pulmonary arteries media hypertrophy and reduced proliferative and inflammatory responses in the lungs. Furthermore, in diseased animals, 2ME (given from day 14 to 28) significantly decreased RVPSP, RV/LV+S ratio and wall thickness, and reduced mortality by 80% (mortality rate: 62.5% vs. 12.5%, MCT vs. MCT+2ME day 14 to 28). This study provides the first evidence that 2ME, a major non-estrogenic, non-carcinogenic metabolite of estradiol, prevents the development and retards the progression of monocrotaline-induced pulmonary hypertension. Further evaluation of 2ME for management of pulmonary arterial hypertension is warranted.
Collapse
Affiliation(s)
- Stevan P Tofovic
- Center for Clinical Pharmacology, University of Pittsburgh School of Medicine, and the VA Pittsburgh Health System, Pittsburgh, PA 15219-3138, USA.
| | | | | | | | | |
Collapse
|
28
|
Tofovic SP, Salah EM, Dubey RK, Melhem MF, Jackson EK. Estradiol Metabolites Attenuate Renal and Cardiovascular Injury Induced by Chronic Nitric Oxide Synthase Inhibition. J Cardiovasc Pharmacol 2005; 46:25-35. [PMID: 15965351 DOI: 10.1097/01.fjc.0000162765.89437.ae] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Our previous studies in rodent models of nephropathy demonstrate that 2-hydroxyestradiol (2HE), an estradiol metabolite with little estrogenic activity, exerts renoprotective effects. In vivo, 2HE is readily converted to 2-methoxyestradiol (2ME), a major estradiol metabolite with no estrogenic activity. The goal of this study was to determine whether 2ME has renal and cardiovascular protective effects in vivo. First, the acute (90 minutes) and chronic (14 days) effects of 2ME (10 microg/kg/h) on blood pressure and renal function were examined in normotensive and spontaneously hypertensive rats (SHR). Second, a rat model of cardiovascular and renal injury induced by chronic nitric oxide synthase inhibition (N-nitro-L-arginine; 40 mg/kg/d; LNNA group) was used to examine the protective effects of estradiol metabolites. Subsets of LNNA-treated rats were administered either 2HE or 2ME (10 microg/kg/h via osmotic minipump; LNNA+2ME and LNNA+2HE groups, respectively. 2-Methoxyestradiol had no acute or chronic effects on blood pressure or renal function in normotensive animals or on hypertension in SHR. Prolonged, 5-week NOS inhibition induced severe cardiovascular and renal disease and high mortality (75%, LNNA group). 2ME, but not 2HE, significantly decreased elevated blood pressure and attenuated the reduction in GFR. 2HE delayed the onset of proteinuria, whereas no proteinuria was detected in the 2-ME group. 2HE and 2ME reduced mortality rate by 66% and 83%, respectively (P < 0.001). In the kidney, 2HE and 2ME abolished LNNA-induced interstitial and glomerular inflammation, attenuated glomerular collagen IV synthesis, and inhibited glomerular and tubular cell proliferation. In the heart, 2HE and 2ME markedly reduced vascular and interstitial inflammation and reduced collagen synthesis and vascular/interstitial cell proliferation. This study provides the first evidence that, in a model of severe cardiovascular and renal injury, 2-methoxyestradiol (a major nonestrogenic estradiol metabolite) exerts renal and cardiovascular protective effects and reduces mortality.
Collapse
Affiliation(s)
- Stevan P Tofovic
- Center for Clinical Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | | | | | | | | |
Collapse
|
29
|
Edgtton KL, Gow RM, Kelly DJ, Carmeliet P, Kitching AR. Plasmin is not protective in experimental renal interstitial fibrosis. Kidney Int 2005; 66:68-76. [PMID: 15200414 DOI: 10.1111/j.1523-1755.2004.00707.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND The plasminogen-plasmin system has potential beneficial or deleterious effects in the context of renal fibrosis. Recent studies have implicated plasminogen activators or their inhibitors in this process. METHODS The development of renal interstitial fibrosis was studied in mice genetically deficient in plasminogen (plg-/- mice) and littermate controls (plg+/+ mice) by inducing unilateral ureteric obstruction (UUO) by ligating the left ureter. RESULTS Collagen accumulation in the kidney was decreased in plg-/- mice at 21 days compared with plg+/+ mice by hydroxyproline assay (plg+/+ 19.0 +/- 1.2 microg collagen/mg tissue, plg-/- 15.6 +/- 0.5 microg collagen/mg tissue, P= 0.04). Macrophage accumulation in plg-/- mice was reduced at 21 days, consistent with a role for plasmin in macrophage recruitment in this model. Myofibroblast accumulation, assessed by the expression of alpha-smooth muscle actin (alpha-SMA), was similar in both groups at both time points. Endogenous plasmin played a role in the activation of transforming growth factor-beta (TGF-beta), as plg-/- mice had lower ratios of betaig-h3:TGF-beta1 mRNA than plg+/+ mice. Matrix metalloproteinase (MMP)-9 activity was unchanged in the absence of plasmin, but MMP-2 activity was decreased. CONCLUSION Plasminogen, the key proenzyme in the plasminogen-plasmin system, does not protect mice from experimental interstitial fibrosis and may have significant pathogenetic effects. These findings, together with other recently published studies in the biology of renal fibrosis, imply that effects of proteins such as plasminogen activator inhibitor-1 (PAI-1), tissue-type plasminogen activator (tPA), and urokinase-type plasminogen activator receptor (uPAR) on renal fibrosis occur independently from the generation of plasmin.
Collapse
Affiliation(s)
- Kristy L Edgtton
- Centre for Inflammatory Diseases, Monash University, Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | | | | | | | | |
Collapse
|
30
|
Zacharia LC, Piché CA, Fielding RM, Holland KM, Allison SD, Dubey RK, Jackson EK. 2-Hydroxyestradiol Is a Prodrug of 2-Methoxyestradiol. J Pharmacol Exp Ther 2004; 309:1093-7. [PMID: 14872091 DOI: 10.1124/jpet.103.062505] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous in vivo studies indicate that 2-hydroxyestradiol (2OHE) attenuates cardiovascular and renal diseases. In vitro studies suggest that the biological effects of 2OHE are mediated by 2-methoxyestradiol (2MEOE) after methylation of 2OHE by catechol-O-methyltransferase (COMT). This study tested the hypothesis that in vivo 2OHE is a prodrug of 2MEOE. We administered to male rats i.v. boluses of either 2OHE or 2MEOE and measured plasma levels of 2OHE and 2MEOE by gas chromatography-mass spectrometry at various time points after drug administration. After administration of 2OHE, plasma levels of 2OHE declined extremely rapidly [t(1/2(1)) = 0.94 min and t(1/2(2)) = 10.2 min] becoming undetectable after 45 min. Concomitant with the disappearance of 2OHE, 2MEOE occurred and then declined [t(1/2(1)) = 7.9 min and t(1/2(2)) = 24.9 min]. The peak concentration and total exposure (area under the curve) for 2OHE were much lower than for 2MEOE. 2OHE had a much higher plasma clearance (CL) and volume of distribution (V(d)) compared with 2MEOE (2OHE: CL = 1215 ml min(-1) kg(-1) and V(d) = 17,875 ml/kg; 2MEOE: CL = 50 ml min(-1) kg(-1) and V(d) = 1760 ml/kg). After administration of 2MEOE, plasma levels of 2MEOE declined [t(1/2(1)) = 2.5 min and t(1/2(2)) = 20.2 min] with a plasma CL of 50 ml min(-1) kg(-1) and a V(d) of 1500 ml/kg. We could not detect 2OHE in plasma from rats receiving 2MEOE. We conclude that the conversion of 2OHE to 2MEOE is so efficient that in terms of 2MEOE exposure, administration of 2OHE is bioequivalent to administration of 2MEOE itself.
Collapse
Affiliation(s)
- Lefteris C Zacharia
- Center for Clinical Pharmacology, 3550 Terrace St., University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Dubey RK, Tofovic SP, Jackson EK. Cardiovascular Pharmacology of Estradiol Metabolites. J Pharmacol Exp Ther 2003; 308:403-9. [PMID: 14657266 DOI: 10.1124/jpet.103.058057] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A discussion of the role of endogenous estradiol metabolites in mediating important biological actions of estradiol is essentially nonexistent in standard textbooks of pharmacology and endocrinology. Indeed, the prevailing view is that all biological effects of estradiol are initiated by binding of estradiol per se to estrogen receptors and that estradiol metabolites are more or less irrelevant. This orthodox view, which is most likely incorrect, is the fundamental premise (an estrogen is an estrogen is an estrogen) underlying the design of important clinical trials such as the Heart and Estrogen/Progestin Replacement Study and the Women's Health Initiative Study. Accumulating data provide convincing evidence that some metabolites of estradiol, the major estrogen secreted by human ovaries, are biologically active and mediate multiple effects on the cardiovascular and renal systems that are largely independent of estrogen receptors. More specifically, metabolites of estradiol, particularly catecholestradiols and methoxyestradiols, induce multiple estrogen receptor-independent actions that protect the heart, blood vessels, and kidneys from disease. These protective effects are mediated in part by the inhibition of the ability of vascular smooth muscle cells, cardiac fibroblasts, and glomerular mesangial cells to migrate, proliferate, and secrete extracellular matrix proteins, as well as by an improvement in vascular endothelial cell function. The purpose of this review is to highlight the cardiovascular and renal pharmacology of catecholestradiols and methoxyestradiols. The take home message is simple: that when it comes to cardiovascular and renal protection, the concept that all estrogenic compounds are created equal may not be true.
Collapse
Affiliation(s)
- Raghvendra K Dubey
- Department of Obstetrics and Gynaecology, Clinic for Endocrinology, University Hospital Zurich, Switzerland
| | | | | |
Collapse
|
32
|
|