1
|
Fujita M. Pulmonary Nontuberculous Mycobacteria Infection in Bronchiectasis: A Narrative Review of Current Status and Future. Health Sci Rep 2025; 8:e70749. [PMID: 40276131 PMCID: PMC12018276 DOI: 10.1002/hsr2.70749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 04/01/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Background and Aims Pulmonary nontuberculous mycobacteria (NTM) infection and bronchiectasis are two distinct respiratory conditions, but bronchiectasis and pulmonary NTM infections are closely associated. NTM can cause bronchiectasis. However, bronchiectasis can create a favorable environment for NTM colonization and exacerbate the progression of NTM. Managing both conditions typically requires a comprehensive approach that addresses infection and the underlying structural lung damage. Methods To perform this review, the author retrieved and assessed relevant articles related to NTM and bronchiectasis that have been published to date from databases, including PubMed/MEDLINE, Scopus, and Google Scholar. Results In this review, the close relationship between pulmonary NTM and bronchiectasis is described from the viewpoints of diagnosis, epidemiology, Pseudomonas aeruginosa, host susceptibility, females and NTM, and treatment. Conclusion Timely diagnosis and management of NTM infections, especially in individuals with underlying risk factors, are essential to prevent disease progression and improve the quality of life of affected individuals.
Collapse
Affiliation(s)
- Masaki Fujita
- Department of Respiratory Medicine, Faculty of MedicineFukuoka University Hospital, Fukuoka UniversityFukuokaJapan
| |
Collapse
|
2
|
Gutiérrez-Brito JA, Lomelí-Nieto JÁ, Muñoz-Valle JF, Oregon-Romero E, Corona-Angeles JA, Hernández-Bello J. Sex hormones and allergies: exploring the gender differences in immune responses. FRONTIERS IN ALLERGY 2025; 5:1483919. [PMID: 39840271 PMCID: PMC11747284 DOI: 10.3389/falgy.2024.1483919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/22/2024] [Indexed: 01/23/2025] Open
Abstract
Allergies are closely associated with sex-related hormonal variations that influence immune function, leading to distinct symptom profiles. Similar sex-based differences are observed in other immune disorders, such as autoimmune diseases. In allergies, women exhibit a higher prevalence of atopic conditions, such as allergic asthma and eczema, in comparison to men. However, age-related changes play a significant role because men have a higher incidence of allergies until puberty, and then comes a switch ratio of prevalence and severity in women. Investigations into the mechanisms of how the hormones influence the development of these diseases are crucial to understanding the molecular, cellular, and pathological aspects. Sex hormones control the reproductive system and have several immuno-modulatory effects affecting immune cells, including T and B cell development, antibody production, lymphoid organ size, and lymphocyte death. Moreover, studies have suggested that female sex hormones amplify memory immune responses, which may lead to an excessive immune response impacting the pathogenesis, airway hyperresponsiveness, inflammation of airways, and mucus production of allergic diseases. The evidence suggests that estrogens enhance immune humoral responses, autoimmunity, mast cell reactivity, and delayed IV allergic reactions, while androgens, progesterone, and glucocorticoids suppress them. This review explores the relationship between sex hormones and allergies, including epidemiological data, experimental findings, and insights from animal models. We discuss the general properties of these hormones, their effects on allergic processes, and clinical observations and therapeutic results. Finally, we describe hypersensitivity reactions to these hormones.
Collapse
Affiliation(s)
| | | | | | | | | | - Jorge Hernández-Bello
- Research Institute of Biomedical Sciences, University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico
| |
Collapse
|
3
|
Agnoletto A, Brisken C. Hormone Signaling in Breast Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:279-307. [PMID: 39821031 DOI: 10.1007/978-3-031-70875-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Hormones control normal breast development and function. They also impinge on breast cancer (BC) development and disease progression in direct and indirect ways. The major ovarian hormones, estrogens and progesterone, have long been established as key regulators of mammary gland development in rodents and linked to human disease. However, their roles have been difficult to disentangle because they act on multiple tissues and can act directly and indirectly on different cell types in the breast, and their receptors interact at different levels within the target cell. Estrogens are well-recognized drivers of estrogen receptor-positive (ER+) breast cancers, and the ER is successfully targeted in ER+ disease. The role of progesterone receptor (PR) as a potential target to be activated or inhibited is debated, and androgen receptor (AR) signaling has emerged as a potentially interesting pathway to target on the stage.In this chapter, we discuss hormone signaling in normal breast development and in cancer, with a specific focus on the key sex hormones: estrogen, progesterone, and testosterone. We will highlight the complexities of endocrine control mechanisms at the organismal, tissue, cellular, and molecular levels. As we delve into the mechanisms of action of hormone receptors, their interplay and their context-dependent roles in breast cancer will be discussed. Drawing insights from new preclinical models, we will describe the lessons learned and the current challenges in understanding hormone action in breast cancer.
Collapse
Affiliation(s)
- Andrea Agnoletto
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Cathrin Brisken
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
4
|
Zhu G, Li J, Xu J, Guo G, Zhu F. Identification of Potential Drug Targets for the Treatment of Severe Burn Wounds from a Multi-Omics Perspective. J Inflamm Res 2024; 17:11275-11289. [PMID: 39720696 PMCID: PMC11668339 DOI: 10.2147/jir.s496429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/23/2024] [Indexed: 12/26/2024] Open
Abstract
Purpose Severe burns result in significant skin damage, impairing its primary role as an infection barrier and presenting substantial treatment challenges. Despite improvements in the treatment of burn patients due to advancements in materials and techniques, there remains a need for novel therapeutic approaches to enhance burn prognosis further. Patients and Methods Several types of genomic methods are used in this study, such as differential gene expression analysis, weighted gene co-expression network analysis (WGCNA), machine learning, and Mendelian randomization (MR), to find genes that are linked to severe burns and create a diagnostic nomogram to see how well these genes can predict severe burns. Drug prediction was conducted using the DsigDB database, and molecular docking was used to validate the pharmacological value of drug targets. The effects of genes and drugs on burn wounds were validated through Western Blot (WB) and cell scratch assays. Results In patients with severe burns, multi-omics analysis revealed increased CYP19A1 expression. In severe burn cell models, WB further confirmed the elevated expression of CYP19A1. Drug prediction indicated that mevastatin binds effectively to the CYP19A1 gene expression protein. The healing area of scalded HaCat cells was much bigger after 24 hours of mevastatin treatment compared to the scald-only group, as shown by cell scratch assays after 24 and 48 hours. Conclusion This study innovatively integrates multi-omics approaches into burn wound research, uncovering for the first time that mevastatin promotes burn wound healing by downregulating CYP19A1 expression. This discovery may provide a new foundation for developing burn wound therapeutics and potentially reduce drug development costs.
Collapse
Affiliation(s)
- Gehua Zhu
- Medical Centre of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Jiaqi Li
- Department of Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, People’s Republic of China
| | - Jiamin Xu
- Medical Centre of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Guanghua Guo
- Medical Centre of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Feng Zhu
- Department of Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, People’s Republic of China
| |
Collapse
|
5
|
Kropp DR, Hodes GE. Sex differences in depression: An immunological perspective. Brain Res Bull 2023; 196:34-45. [PMID: 36863664 DOI: 10.1016/j.brainresbull.2023.02.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/05/2023] [Accepted: 02/27/2023] [Indexed: 03/04/2023]
Abstract
Depression is a heterogenous disorder with symptoms that present differently across individuals. In a subset of people depression is associated with alterations of the immune system that may contribute to disorder onset and symptomology. Women are twice as likely to develop depression and on average have a more sensitive adaptive and innate immune system when compared to men. Sex differences in pattern recognition receptors (PRRs), release of damage-associated molecular patterns (DAMPs), cell populations, and circulating cytokines play a critical role in inflammation onset. Sex differences in innate and adaptive immunity change the response of and repair to damage caused by dangerous pathogens or molecules in the body. This article reviews the evidence for sex specific immune responses that contribute to the sex differences in symptoms of depression that may account for the higher rate of depression in women.
Collapse
Affiliation(s)
- Dawson R Kropp
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Georgia E Hodes
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
6
|
Dong Y, Li X, Li Z, Zhu Y, Wei Z, He J, Cheng H, Yang A, Chen F. Effects of inactivated SARS-CoV-2 vaccination on male fertility: A retrospective cohort study. J Med Virol 2023; 95:e28329. [PMID: 36415120 DOI: 10.1002/jmv.28329] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/27/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022]
Abstract
Numerous studies have revealed severe damage to male fertility from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, raising concerns about the potential adverse impact on reproductive function of the coronavirus disease 2019 (COVID-19) vaccine developed based on the virus. Interestingly, there are several researchers who have studied the impact of the COVID-19 mRNA vaccine since then but have come up with conflicting results. As a near-ideal candidate for mass immunization programs, inactivated SARS-CoV-2 vaccine has been widely used in many countries, particularly in less wealthy nations. However, little is known about its effect on male fertility. Here, we conducted a retrospective cohort study at a single large center for reproductive medicine in China between December 2021 and August 2022. Five hundred and nineteen fertile men with no history of laboratory-confirmed COVID-19 were included and categorized into four groups based on their vaccination status: unvaccinated group (n = 168), one-dose vaccinated group (n = 8), fully vaccinated group (n = 183), and booster group (n = 160). All of them underwent a semen analysis and most had serum sex hormone levels tested. There were no significant differences in all semen parameters and sex hormone levels between the unvaccinated group and either vaccinated group. To account for possible vaccination-to-test interval-specific changes, sub-analyses were performed for two interval groups: ≤90 and >90 days. As expected, most of the semen parameters and sex hormone levels remained unchanged between the control and vaccinated groups. However, participants in vaccinated group (≤90 days) have decreased total sperm motility and increased follicle-stimulating hormone level compared with the ones in unvaccinated group. Moreover, some trends similar to those found during COVID-19 infection and recovery were observed in our study. Fortunately, all values are within the normal range. In addition, vaccinated participants reported few adverse reactions. No special medical intervention was required, and no serious adverse reactions happened. Our study suggests that inactivated SARS-CoV-2 vaccination does not impair male fertility, possibly due to the low frequency of adverse effects. This information reassures young male population who got this vaccine worldwide, and helps guide future vaccination efforts.
Collapse
Affiliation(s)
- Yehao Dong
- Center for Reproductive Medicine, Affiliated Hospital of Jining Medical University, Jining, China
| | - Xiaoyun Li
- Center for Reproductive Medicine, Affiliated Hospital of Jining Medical University, Jining, China
| | - Zewu Li
- Center for Reproductive Medicine, Affiliated Hospital of Jining Medical University, Jining, China
| | - Yunting Zhu
- Department of Physiology, Jining Medical University, Jining, China
| | - Zichun Wei
- Department of Physiology, Jining Medical University, Jining, China
| | - Jiarui He
- Department of Physiology, Jining Medical University, Jining, China
| | - Hongju Cheng
- Department of Physiology, Jining Medical University, Jining, China
| | - Aijun Yang
- Center for Reproductive Medicine, Affiliated Hospital of Jining Medical University, Jining, China
| | - Fei Chen
- Department of Physiology, Jining Medical University, Jining, China
| |
Collapse
|
7
|
Collins MK, McCutcheon CR, Petroff MG. Impact of Estrogen and Progesterone on Immune Cells and Host–Pathogen Interactions in the Lower Female Reproductive Tract. THE JOURNAL OF IMMUNOLOGY 2022; 209:1437-1449. [DOI: 10.4049/jimmunol.2200454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/05/2022] [Indexed: 11/05/2022]
|
8
|
True H, Blanton M, Sureshchandra S, Messaoudi I. Monocytes and macrophages in pregnancy: The good, the bad, and the ugly. Immunol Rev 2022; 308:77-92. [PMID: 35451089 DOI: 10.1111/imr.13080] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/08/2022] [Indexed: 12/12/2022]
Abstract
A successful human pregnancy requires precisely timed adaptations by the maternal immune system to support fetal growth while simultaneously protecting mother and fetus against microbial challenges. The first trimester of pregnancy is characterized by a robust increase in innate immune activity that promotes successful implantation of the blastocyst and placental development. Moreover, early pregnancy is also a state of increased vulnerability to vertically transmitted pathogens notably, human immunodeficiency virus (HIV), Zika virus (ZIKV), SARS-CoV-2, and Listeria monocytogenes. As gestation progresses, the second trimester is marked by the establishment of an immunosuppressive environment that promotes fetal tolerance and growth while preventing preterm birth, spontaneous abortion, and other gestational complications. Finally, the period leading up to labor and parturition is characterized by the reinstatement of an inflammatory milieu triggering childbirth. These dynamic waves of carefully orchestrated changes have been dubbed the "immune clock of pregnancy." Monocytes in maternal circulation and tissue-resident macrophages at the maternal-fetal interface play a critical role in this delicate balance. This review will summarize the current data describing the longitudinal changes in the phenotype and function of monocyte and macrophage populations in healthy and complicated pregnancies.
Collapse
Affiliation(s)
- Heather True
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA.,Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - Madison Blanton
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA.,Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | | | - Ilhem Messaoudi
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
9
|
Progesterone and Inflammatory Response in the Oviduct during Physiological and Pathological Conditions. Cells 2022; 11:cells11071075. [PMID: 35406639 PMCID: PMC8997425 DOI: 10.3390/cells11071075] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 11/25/2022] Open
Abstract
Progesterone has been shown to be a potent suppressor of several inflammatory pathways. During pregnancy, progesterone levels increase, allowing for normal pregnancy establishment and maintenance. The dysregulation of progesterone, as well as inflammation, leads to poor pregnancy outcomes. However, it is unclear how progesterone imbalance could impact inflammatory responses in the oviduct and subsequently result in early pregnancy loss. Therefore, in this review, we describe the role of progesterone signaling in regulating the inflammatory response, with a focus on the oviduct and pathological conditions in the Fallopian tubes.
Collapse
|
10
|
Bharath LP, Regan T, Conway R. Regulation of Immune Cell Function by Nicotinamide Nucleotide Transhydrogenase. Am J Physiol Cell Physiol 2022; 322:C666-C673. [PMID: 35138175 PMCID: PMC8977145 DOI: 10.1152/ajpcell.00607.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Redox homeostasis is elemental for the normal physiology of all cell types. Cells use multiple mechanisms to regulate the redox balance tightly. The onset and progression of many metabolic and aging-associated diseases occur due to the dysregulation of redox homeostasis. Thus, it is critical to identify and therapeutically target mechanisms that precipitate abnormalities in redox balance. Reactive oxygen species (ROS) produced within the immune cells regulate homeostasis, hyperimmune and hypoimmune cell responsiveness, apoptosis, immune response to pathogens, and tumor immunity. Immune cells have both cytosolic and organelle-specific redox regulatory systems to maintain appropriate levels of ROS. Nicotinamide nucleotide transhydrogenase (NNT) is an essential mitochondrial redox regulatory protein. Dysregulation of NNT function prevents immune cells from mounting an adequate immune response to pathogens, promotes a chronic inflammatory state associated with aging and metabolic diseases, and initiates conditions related to a dysregulated immune system such as autoimmunity. While many studies have reported on NNT in different cell types, including cancer cells, relatively few studies have explored NNT in immune cells. This review provides an overview of NNT and focuses on the current knowledge of NNT in the immune cells.
Collapse
Affiliation(s)
- Leena P Bharath
- Department of Nutrition and Public Health, Merrimack College, North Andover, Massachusetts, United States
| | - Thomas Regan
- Department of Nutrition and Public Health, Merrimack College, North Andover, Massachusetts, United States
| | - Rachel Conway
- Department of Nutrition and Public Health, Merrimack College, North Andover, Massachusetts, United States
| |
Collapse
|
11
|
Groh LA, Verel DE, van der Heijden CDCC, Matzaraki V, Moorlag SJCFM, de Bree LC, Koeken VACM, Mourits VP, Keating ST, van Puffelen JH, Joosten LAB, Netea MG, Riksen NP. Immune modulatory effects of progesterone on oxLDL-induced trained immunity in monocytes. J Leukoc Biol 2022; 112:279-288. [PMID: 35040511 PMCID: PMC9544104 DOI: 10.1002/jlb.3ab1220-846r] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Atherosclerotic cardiovascular diseases (CVD) are among the leading causes of death in the world. Monocyte‐derived macrophages are key players in the pathophysiology of atherosclerosis. Innate immune memory following exposure of monocytes to atherogenic compounds, such as oxidized low‐density lipoproteins (oxLDL), termed trained immunity, can contribute to atherogenesis. The current study aimed to elucidate intracellular mechanisms of oxLDL‐induced trained immunity. Using untargeted intracellular metabolomics in isolated human primary monocytes, we show that oxLDL‐induced trained immunity results in alterations in the balance of intracellular steroid hormones in monocytes. This was reflected by a decrease in extracellular progesterone concentrations following LPS stimulation. To understand the potential effects of steroid hormones on trained immunity, monocytes were costimulated with oxLDL and the steroid hormones progesterone, hydrocortisone, dexamethasone, β‐estradiol, and dihydrotestosterone. Progesterone showed a unique ability to attenuate the enhanced TNFα and IL‐6 production following oxLDL‐induced trained immunity. Single nucleotide polymorphisms in the nuclear glucocorticoid, progesterone, and mineralocorticoid receptor were shown to correlate with ex vivo oxLDL‐induced trained immunity in 243 healthy volunteers. Pharmacologic inhibition experiments revealed that progesterone exerts the suppression of TNFα in trained immunity via the nuclear glucocorticoid and mineralocorticoid receptors. Our data show that progesterone has a unique ability to suppress oxLDL‐induced trained immunity. We hypothesize that this effect might contribute to the lower incidence of CVD in premenopausal women.
Collapse
Affiliation(s)
- Laszlo A Groh
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dagmar E Verel
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Charlotte D C C van der Heijden
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vasiliki Matzaraki
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Simone J C F M Moorlag
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - L Charlotte de Bree
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Valerie A C M Koeken
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Computational Biology for Individualised Infection Medicine, Centre for Individualised Infection Medicine (CiiM) & TWINCORE, Joint Ventures Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Vera P Mourits
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Samuel T Keating
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jelmer H van Puffelen
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Niels P Riksen
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
12
|
Dutta S, Sandhu N, Sengupta P, Alves MG, Henkel R, Agarwal A. Somatic-Immune Cells Crosstalk In-The-Making of Testicular Immune Privilege. Reprod Sci 2021; 29:2707-2718. [PMID: 34580844 DOI: 10.1007/s43032-021-00721-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/22/2021] [Indexed: 11/27/2022]
Abstract
Immunological infertility contributes significantly to the etiology of idiopathic male infertility. Shielding the spermatogenic cells from systemic immune responses is fundamental to secure normal production of spermatozoa. The body's immune system is tuned with the host self-components since the early postnatal period, while sperm first develops during puberty, thus rendering spermatogenic proteins as 'non-self' or 'antigenic.' Development of antibodies to these antigens elicits autoimmune responses affecting sperm motility, functions, and fertility. Therefore, the testes need to establish a specialized immune-privileged microenvironment to protect the allogenic germ cells by orchestration of various testicular cells and resident immune cells. This is achieved through sequestration of antigenic germ cells by blood-testis barrier and actions of various endocrine, paracrine, immune-suppressive, and immunomodulatory mechanisms. The various mechanisms are very complex and need conceptual integration to disclose the exact physiological scenario, and to facilitate detection and management of immunogenic infertility caused by disruption of testicular immune regulation. The present review aims to (a) discuss the components of testicular immune privilege; (b) explain testicular somatic and immune cell interactions in establishing and maintaining the testicular immune micro-environment; and (c) illustrate the integration of multiple mechanisms involved in the control of immune privilege of the testis.
Collapse
Affiliation(s)
- Sulagna Dutta
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Jenjarom, Selangor , Malaysia
| | - Narpal Sandhu
- Molecular and Cellular Biology, University of California, Berkeley, CA, USA
| | - Pallav Sengupta
- Department of Physiology, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jenjarom, Selangor , Malaysia
| | - Marco G Alves
- Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Ralf Henkel
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Medical Bioscience, University of the Western Cape, Bellville, South Africa
- LogixX Pharma, Theale, Berkshire, UK
| | - Ashok Agarwal
- American Center for Reproductive Medicine, Cleveland Clinic, Mail Code X-11, 10681 Carnegie Avenue, Cleveland, OH, 44195, USA.
| |
Collapse
|
13
|
Characterization of Membrane-Associated Progesterone Receptor Component-2 (MAPRC2) from Trichinella spiralis and Its Interaction with Progesterone and Mifepristone. Vaccines (Basel) 2021; 9:vaccines9080934. [PMID: 34452060 PMCID: PMC8402905 DOI: 10.3390/vaccines9080934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 11/18/2022] Open
Abstract
Trichinellosis is a foodborne zoonotic disease caused by Trichinella spp., including Trichinella spiralis. In the present study, T. spiralis membrane-associated progesterone receptor component-2 (Ts-MAPRC2) gene was cloned and characterized using protein sequencing analysis. Furthermore, the expression, purification, immunoblot assay, binding ability with progesterone antibody, and immunofluorescence assay were performed. A direct effect of progesterone (P4) and mifepristone (RU486) on the Ts-MAPRC2 gene was determined using in vitro cell culture that showed different expression levels at all developmental stages (muscle larvae (ML), female adult worm (F-AL), male adult worm (M-AL), and newborn larvae (NBL)). Subsequently, the in vitro phenotypic effects of P4, RU486, and rTs-MAPRC2-Ab on F-AL and ML stages were measured. Later, the in vivo phenotypic effect and relative mRNA expression of mifepristone on the F-AL stage were studied. Our results revealed that the Ts-MAPRC2 gene is critical to maintaining pregnancy in the female adult worm (F-AL) of T. spiralis. The 300 ng/mL of P4 and 100 ng/mL of RU486 showed downregulation of the Ts-MAPRC2 gene in F-AL (p ≤ 0.05). This plays an important role in abortion and possibly decreases the worm burden of T. spiralis in the host. Only 30 ng/mL P4 showed significant upregulation in F-AL (p ≤ 0.05). The current study provides new insights regarding the antihormone (P4 and RU486) drug design and vaccine therapy of recombinant (rTs-MAPRC2) protein as well as their combined effects to control T. spiralis infection.
Collapse
|
14
|
Glodić G, Samaržija M, Sabol I, Bulat Kardum L, Carević Vladić V, Džubur F, Jakopović M, Kuhtić I, Marušić A, Mihelčić D, Popović Grle S, Strelec D, Šklebar T, Šola AM, Žmak L, van Ingen J, Janković Makek M. Risk factors for nontuberculous mycobacterial pulmonary disease (NTM-PD) in Croatia. Wien Klin Wochenschr 2021; 133:1195-1200. [PMID: 34402990 DOI: 10.1007/s00508-021-01923-x] [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: 01/04/2021] [Accepted: 07/09/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND The incidence, geographical distribution and clinical relevance of different nontuberculous mycobacteria (NTM) in Croatia are well described. There are few data on the risk factors for developing NTM pulmonary disease (NTM-PD) in this setting. METHODS We conducted a retrospective cohort study on all Croatian residents with NTM isolated from respiratory samples in the period from 2006 to 2015 with follow-up to 2018. The American Thoracic Society/Infectious Diseases Society of America (ATS/IDSA) guidelines were used to establish NTM-PD diagnosis. Clinical, radiological and treatment data were collected from hospital records. RESULTS Risk analysis calculations were made on the 439 isolation episodes that were classified as definitive NTM-PD (n = 137) or no disease (n = 302). Female gender, presence of bronchiectasis, low BMI and long-term systemic corticosteroid treatment were independent risk factors associated with NTM-PD. Hemoptysis and malaise were presenting symptoms independently associated with NTM-PD. Chronic obstructive pulmonary disease (COPD) and low/moderate dose inhaled corticosteroid (ICS) treatment were not associated with NTM-PD. High dose ICS treatment was a significant risk factor for developing NTM-PD (aOR = 4.73, CI 1.69-13.23 p = 0.003). CONCLUSION The NTM-PD patients in Croatia are similar to those in other published cohorts in terms of their characteristics and risk factors. The significant dose-dependent association between ICS use and NTM-PD adds to the body of evidence suggesting that high dose ICS use is associated with NTM-PD.
Collapse
Affiliation(s)
- Goran Glodić
- Clinic for Respiratory diseases Jordanovac, University Hospital Centre Zagreb, Zagreb, Croatia.
| | - Miroslav Samaržija
- Clinic for Respiratory diseases Jordanovac, University Hospital Centre Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ivan Sabol
- Division of Molecular Medicine, Laboratory od Molecular Virology and Bacteriology, Ruder Boskovic Institute, Zagreb, Croatia
| | - Ljiljana Bulat Kardum
- Department for Respiratory Diseases, Clinical Hospital Center Rijeka, Rijeka, Croatia
| | | | - Feđa Džubur
- Clinic for Respiratory diseases Jordanovac, University Hospital Centre Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Marko Jakopović
- Clinic for Respiratory diseases Jordanovac, University Hospital Centre Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ivana Kuhtić
- Clinic for Radiology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Ante Marušić
- Clinic for Radiology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Dina Mihelčić
- Clinic for Respiratory diseases Jordanovac, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Sanja Popović Grle
- Clinic for Respiratory diseases Jordanovac, University Hospital Centre Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dražen Strelec
- Hospital for pulmonary disease and TBC Klenovnik, Klenovnik, Croatia
| | | | - Ana Marija Šola
- Special Hospital for Respiratory Diseases Rockefellerova, Zagreb, Croatia
| | - Ljiljana Žmak
- School of Medicine, University of Zagreb, Zagreb, Croatia
- National Mycobacteria Reference Laboratory, Croatian Institute of Public Health, Zagreb, Croatia
| | - Jakko van Ingen
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmengen, The Netherlands
| | - Mateja Janković Makek
- Clinic for Respiratory diseases Jordanovac, University Hospital Centre Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
15
|
Gay L, Melenotte C, Lakbar I, Mezouar S, Devaux C, Raoult D, Bendiane MK, Leone M, Mège JL. Sexual Dimorphism and Gender in Infectious Diseases. Front Immunol 2021; 12:698121. [PMID: 34367158 PMCID: PMC8339590 DOI: 10.3389/fimmu.2021.698121] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022] Open
Abstract
Epidemiological studies and clinical observations show evidence of sexual dimorphism in infectious diseases. Women are at less risk than men when it comes to developing most infectious diseases. However, understanding these observations requires a gender approach that takes into account an analysis of both biological and social factors. The host’s response to infection differs in males and females because sex differences have an impact on hormonal and chromosomal control of immunity. Estradiol appears to confer protective immunity, while progesterone and testosterone suppress anti-infectious responses. In addition, genetic factors, including those associated with sex chromosomes, also affect susceptibility to infections. Finally, differences in occupational activities, lifestyle, and comorbidities play major roles in exposure to pathogens and management of diseases. Hence, considering sexual dimorphism as a critical variable for infectious diseases should be one of the steps taken toward developing personalized therapeutic approaches.
Collapse
Affiliation(s)
- Laetitia Gay
- Aix-Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | - Cléa Melenotte
- Aix-Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France.,Aix-Marseille Univ, INSERM, IRD, SESSTIM, Economy and Social Science, Health Care Systems and Societies, Marseille, France
| | - Ines Lakbar
- Department of Anaesthesia and Intensive Care, Hôpital Nord, Aix-Marseille Univ, APHM, Marseille, France
| | - Soraya Mezouar
- Aix-Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | - Christian Devaux
- Aix-Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | - Didier Raoult
- Aix-Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | - Marc-Karim Bendiane
- Aix-Marseille Univ, INSERM, IRD, SESSTIM, Economy and Social Science, Health Care Systems and Societies, Marseille, France
| | - Marc Leone
- Aix-Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France.,Department of Anaesthesia and Intensive Care, Hôpital Nord, Aix-Marseille Univ, APHM, Marseille, France
| | - Jean-Louis Mège
- Aix-Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| |
Collapse
|
16
|
Correa-Martínez CL, Schuler F, Kampmeier S. Sex differences in vancomycin-resistant enterococci bloodstream infections-a systematic review and meta-analysis. Biol Sex Differ 2021; 12:36. [PMID: 34001270 PMCID: PMC8130152 DOI: 10.1186/s13293-021-00380-5] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
Background Vancomycin-resistant enterococci (VRE) have emerged in the healthcare setting worldwide. Infections with these pathogens, i.e., bloodstream infections (BSI), are accompanied with an impaired patient outcome. Diverse factors comprising patient characteristics, therapeutic strategies, and infection control measures are positively or negatively associated with VRE BSI occurrence. However, whether sex-specific differences influence the frequency of VRE BSI is yet unknown. The aim of this systematic review was to comprehensively summarize and analyze sex prevalence in VRE BSI patients. Main text A systematic search for relevant articles was conducted in PubMed and Web of Science. After screening for eligibility, data extraction from included articles and risk of bias assessment were processed. The prevalence of male/female sex in VRE BSI patients and 95% CI were calculated for each study and summarized as pooled estimated effect. In total, nine articles met the inclusion criteria. Risk of bias assessment resulted in low (six studies) to moderate bias (three studies). The pooled prevalence of male patients suffering from VRE BSI was 59% resulting in a 1.4 male/female prevalence ratio. Conclusions Current literature suggests sex differences with male preference (59%) in the distribution of VRE BSI cases. Further primary studies should address the question of male-specific factors favoring the enhanced frequency of VRE BSI.
Collapse
Affiliation(s)
- Carlos L Correa-Martínez
- Institute of Hygiene, University Hospital Münster, Robert-Koch-Strasse 41, 48149, Münster, Germany
| | - Franziska Schuler
- Institute of Medical Microbiology, University Hospital Münster, Domagkstrasse 10, 48149, Münster, Germany
| | - Stefanie Kampmeier
- Institute of Hygiene, University Hospital Münster, Robert-Koch-Strasse 41, 48149, Münster, Germany.
| |
Collapse
|
17
|
Dziedzic A, Saluk-Bijak J, Miller E, Niemcewicz M, Bijak M. The Impact of SARS-CoV-2 Infection on the Development of Neurodegeneration in Multiple Sclerosis. Int J Mol Sci 2021; 22:1804. [PMID: 33670394 PMCID: PMC7918534 DOI: 10.3390/ijms22041804] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 02/07/2023] Open
Abstract
The novel coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), remains a global challenge. Currently, there is some information on the consequences of COVID-19 infection in multiple sclerosis (MS) patients, as it is a newly discovered coronavirus, but its far-reaching effects on participation in neurodegenerative diseases seem to be significant. Recent cases reports showed that SARS-CoV-2 may be responsible for initiating the demyelination process in people who previously had no symptoms associated with any nervous system disorders. It is presently known that infection of SARS-CoV-2 evokes cytokine storm syndrome, which may be one of the factors leading to the acute cerebrovascular disease. One of the substantial problems is the coexistence of cerebrovascular disease and MS in an individual's life span. Epidemiological studies showed an enhanced risk of death rate from vascular disabilities in MS patients of approximately 30%. It has been demonstrated that patients with severe SARS-CoV-2 infection usually show increased levels of D-dimer, fibrinogen, C-reactive protein (CRP), and overactivation of blood platelets, which are essential elements of prothrombotic events. In this review, the latest knowledge gathered during an ongoing pandemic of SARS-CoV-2 infection on the neurodegeneration processes in MS is discussed.
Collapse
Affiliation(s)
- Angela Dziedzic
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland;
| | - Joanna Saluk-Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland;
| | - Elzbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland;
| | - Marcin Niemcewicz
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (M.N.); (M.B.)
| | - Michal Bijak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (M.N.); (M.B.)
| |
Collapse
|
18
|
Functional regulation of decidual macrophages during pregnancy. J Reprod Immunol 2020; 143:103264. [PMID: 33360717 DOI: 10.1016/j.jri.2020.103264] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/22/2020] [Accepted: 11/25/2020] [Indexed: 12/16/2022]
Abstract
A successful pregnancy requires that the maternal immune system recognizes and tolerates the semi-allogeneic fetus without compromising the capability of protecting both mother and fetus from various pathogens. Decidual macrophages present unique phenotypes to play a key role in the establishment of the immunological aspects of maternal-fetal interaction. Dysfunction of decidual macrophages gives rise to pregnancy complications such as preeclampsia, recurrent spontaneous miscarriage, preterm labor and fetal growth restriction. Here, we reviewed the latest knowledge on the origin, differentiation, unique phenotype and function of macrophages in normal pregnancy and in pregnancy complications. We mainly focused on the significant roles of decidual macrophages in the process of extravillous trophoblast invasion, spiral arterial remodeling, decidual stromal cells cultivation and immune tolerance maintenance in normal pregnancy, and their pathological roles in pregnancy-related complications, offering more integrated information in maternal-fetal immunity.
Collapse
|
19
|
Gadsby JE, Frandsen S, Chang J, Celestino B, Tucker E, Poole DH. Progesterone inhibits cytokine/TNF-α production by porcine CL macrophages via the genomic progesterone receptor. Domest Anim Endocrinol 2020; 72:106426. [PMID: 32244110 DOI: 10.1016/j.domaniend.2019.106426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/18/2019] [Accepted: 12/06/2019] [Indexed: 11/26/2022]
Abstract
In pigs, luteolytic sensitivity to PGF-2α (=LS) is delayed until d 13 of the estrous cycle. While the control of LS is unknown, it is temporally associated with macrophage (MAC; which secretes tumor necrosis factor [TNF]-α) infiltration into the corpora lutea (CL), and previous studies have shown that TNF-α induces LS in porcine luteal cells (LCs) in culture. This study was designed to explore the control of LS by CL macrophage (CL MAC)/TNF-α by progesterone (P4), and to examine the hypothesis that P4 acting via the genomic P4 receptor (PGR) inhibits CL MAC TNF-α and thus plays a key role in regulating LS during the pig estrous cycle. In experiment 1, the effects of LCs on CL MAC cytokine/TNF-α mRNA expression in co-culture were examined (MID cycle; ~d 7-12; no LS); results showed that LC was inhibitory to cytokine/TNF-α. In experiment 2, the effects of P4 or R5020 (PGR-agonist) on CL MAC cytokine/TNF-α mRNA expression were examined (MID cycle; ~d 7-12; no LS); results showed that both P4 and R5020 dose-dependently inhibited TNF-α. In experiment 3, CL MACs were isolated from CL at MID (~d 7-12; no LS) and LATE (~d 13-18; + LS) cycle, and TNF-α/PGR mRNA measured. Results indicated that while TNF-α mRNA was 4.2-fold greater in CL MACs from LATE vs MID cycle, PGR mRNA was 4.5-fold greater in CL MACs from MID vs LATE cycle. These data support our hypothesis and suggest that progesterone, acting via PGR, plays a critical physiological role in the control of TNF-α production by CL MACs and LS during the pig estrous cycle.
Collapse
Affiliation(s)
- J E Gadsby
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.
| | - S Frandsen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - J Chang
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - B Celestino
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - E Tucker
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - D H Poole
- Department of Animal Science, College of Agricultural and Life Sciences, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
20
|
Nathan K, Lu LY, Lin T, Pajarinen J, Jämsen E, Huang JF, Romero-Lopez M, Maruyama M, Kohno Y, Yao Z, Goodman SB. Precise immunomodulation of the M1 to M2 macrophage transition enhances mesenchymal stem cell osteogenesis and differs by sex. Bone Joint Res 2019; 8:481-488. [PMID: 31728188 PMCID: PMC6825050 DOI: 10.1302/2046-3758.810.bjr-2018-0231.r2] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES Up to 10% of fractures result in undesirable outcomes, for which female sex is a risk factor. Cellular sex differences have been implicated in these different healing processes. Better understanding of the mechanisms underlying bone healing and sex differences in this process is key to improved clinical outcomes. This study utilized a macrophage-mesenchymal stem cell (MSC) coculture system to determine: 1) the precise timing of proinflammatory (M1) to anti-inflammatory (M2) macrophage transition for optimal bone formation; and 2) how such immunomodulation was affected by male versus female cocultures. METHODS A primary murine macrophage-MSC coculture system was used to demonstrate the optimal transition time from M1 to M2 (polarized from M1 with interleukin (IL)-4) macrophages to maximize matrix mineralization in male and female MSCs. Outcome variables included Alizarin Red staining, alkaline phosphatase (ALP) activity, and osteocalcin protein secretion. RESULTS We found that 96 hours of M1 phenotype in male cocultures allowed for maximum matrix mineralization versus 72 hours in female cocultures. ALP activity and osteocalcin secretion were also enhanced with the addition of IL-4 later in male versus female groups. The sex of the cells had a statistically significant effect on the optimal IL-4 addition time to maximize osteogenesis. CONCLUSION These results suggest that: 1) a 72- to 96-hour proinflammatory environment is critical for optimal matrix mineralization; and 2) there are immunological differences in this coculture environment due to sex. Optimizing immunomodulation during fracture healing may enhance and expedite the bone regeneration response. These findings provide insight into precise immunomodulation for enhanced bone healing that is sex-specific.Cite this article: K. Nathan, L. Y. Lu, T. Lin, J. Pajarinen, E. Jämsen, J-F. Huang, M. Romero-Lopez, M. Maruyama, Y. Kohno, Z. Yao, S. B. Goodman. Precise immunomodulation of the M1 to M2 macrophage transition enhances mesenchymal stem cell osteogenesis and differs by sex. Bone Joint Res 2019;8:481-488. DOI: 10.1302/2046-3758.810.BJR-2018-0231.R2.
Collapse
Affiliation(s)
- Karthik Nathan
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Laura Y Lu
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Jukka Pajarinen
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Eemeli Jämsen
- Department of Medicine, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Jhih-Fong Huang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Monica Romero-Lopez
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Yusuke Kohno
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA; Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
21
|
Zheng H, Guo Q, Duan X, Xu Z, Wang Q. l-arginine inhibited apoptosis of fish leukocytes via regulation of NF-κB-mediated inflammation, NO synthesis, and anti-oxidant capacity. Biochimie 2019; 158:62-72. [DOI: 10.1016/j.biochi.2018.12.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 12/16/2018] [Indexed: 02/07/2023]
|
22
|
Heuerman AC, Hollinger TT, Menon R, Mesiano S, Yellon SM. Cervix Stromal Cells and the Progesterone Receptor A Isoform Mediate Effects of Progesterone for Prepartum Remodeling. Reprod Sci 2019; 26:690-696. [PMID: 30654718 DOI: 10.1177/1933719118820462] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The prepartum transition from a soft to ripening cervix is an inflammatory process that occurs well before birth when systemic progesterone is near peak concentration. This 2-part study first determined that stromal fibroblasts but not macrophages in the cervix have progesterone receptors (PRs). Neither the number of PR cells in cervix sections nor the relative abundance or ratio of nuclear PR isoforms (PR-A/PR-B) were diminished in mice between day 15 of pregnancy and term. Second in mice lacking PR-B ( Pgrtm20mc), the number of cells that expressed the PR-A isoform were maintained during this period of prepartum cervix remodeling. Thus, progesterone effects to sustain pregnancy, as well as soften and ripen the cervix, are mediated by a stable stromal cell population that expresses PR-A and, through interactions with resident macrophages, are likely to mediate inflammatory ripening processes in preparation for birth.
Collapse
Affiliation(s)
- Anne C Heuerman
- 1 Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Trevor T Hollinger
- 1 Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Ramkumar Menon
- 2 Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Sam Mesiano
- 3 Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Steven M Yellon
- 1 Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, USA.,4 Division of Physiology, Department of Basic Sciences, and Departments of Basic Sciences and Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
23
|
Cioni B, Zwart W, Bergman AM. Androgen receptor moonlighting in the prostate cancer microenvironment. Endocr Relat Cancer 2018; 25:R331-R349. [PMID: 29618577 DOI: 10.1530/erc-18-0042] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/04/2018] [Indexed: 01/03/2023]
Abstract
Androgen receptor (AR) signaling is vital for the normal development of the prostate and is critically involved in prostate cancer (PCa). AR is not only found in epithelial prostate cells but is also expressed in various cells in the PCa-associated stroma, which constitute the tumor microenvironment (TME). In the TME, AR is expressed in fibroblasts, macrophages, lymphocytes and neutrophils. AR expression in the TME was shown to be decreased in higher-grade and metastatic PCa, suggesting that stromal AR plays a protective role against PCa progression. With that, the functionality of AR in stromal cells appears to deviate from the receptor's classical function as described in PCa cells. However, the biological action of AR in these cells and its effect on cancer progression remains to be fully understood. Here, we systematically review the pathological, genomic and biological literature on AR actions in various subsets of prostate stromal cells and aim to better understand the consequences of AR signaling in the TME in relation to PCa development and progression.
Collapse
Affiliation(s)
- B Cioni
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - W Zwart
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode InstituteThe Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - A M Bergman
- Division of OncogenomicsThe Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Medical OncologyThe Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
24
|
Noninvasive detection of macrophage activation with single-cell resolution through machine learning. Proc Natl Acad Sci U S A 2018; 115:E2676-E2685. [PMID: 29511099 DOI: 10.1073/pnas.1711872115] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We present a method enabling the noninvasive study of minute cellular changes in response to stimuli, based on the acquisition of multiple parameters through label-free microscopy. The retrieved parameters are related to different attributes of the cell. Morphological variables are extracted from quantitative phase microscopy and autofluorescence images, while molecular indicators are retrieved via Raman spectroscopy. We show that these independent parameters can be used to build a multivariate statistical model based on logistic regression, which we apply to the detection at the single-cell level of macrophage activation induced by lipopolysaccharide (LPS) exposure and compare their respective performance in assessing the individual cellular state. The models generated from either morphology or Raman can reliably and independently detect the activation state of macrophage cells, which is validated by comparison with their cytokine secretion and intracellular expression of molecules related to the immune response. The independent models agree on the degree of activation, showing that the features provide insight into the cellular response heterogeneity. We found that morphological indicators are linked to the phenotype, which is mostly related to downstream effects, making the results obtained with these variables dose-dependent. On the other hand, Raman indicators are representative of upstream intracellular molecular changes related to specific activation pathways. By partially inhibiting the LPS-induced activation using progesterone, we could identify several subpopulations, showing the ability of our approach to identify the effect of LPS activation, specific inhibition of LPS, and also the effect of progesterone alone on macrophage cells.
Collapse
|
25
|
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: 24] [Impact Index Per Article: 3.0] [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
|
26
|
The first data on the infestation of the parti-coloured bat, Vespertilio murinus (Chiroptera, Vespertilionidae), with gamasid mites, Steatonyssus spinosus (Mesostigmata, Gamasina, Macronyssidae). RUSSIAN JOURNAL OF THERIOLOGY 2017. [DOI: 10.15298/rusjtheriol.16.1.06] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
27
|
Watanabe H, Uruma T, Seita I, Oishi T, Watanabe Y, Tsukimori A, Haga Y, Fukushima S, Sato A, Nakamura I, Matsumoto T. Solitary pulmonary caseating granulomas: A 5-year retrospective single-center analysis. Mol Clin Oncol 2017; 6:839-845. [PMID: 28588774 PMCID: PMC5451854 DOI: 10.3892/mco.2017.1244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/10/2017] [Indexed: 11/06/2022] Open
Abstract
Solitary pulmonary caseating granulomas (SPCGs) are a characteristic type of tuberculomas associated with infection with non-tuberculous mycobacteria (NTM) and other microbes; however, their significance remains unclear. The aim of the present study was to describe the clinical characteristics of patients with SPCGs in terms of diagnosis, presence of lung cancer and treatment status. A retrospective analysis of 17 immunocompetent patients with histopathologically confirmed caseating granulomas after undergoing video-assisted thoracoscopic surgery (VATS) was conducted at our center between 2011 and 2015. The patients comprised 10 men and 7 women with a mean age of 59.1±14.4 years. Of the 17 patients, 14 (82.4%) were asymptomatic and the lesions were discovered incidentally. In 2 patients the SPCGs were accompanied by a small satellite nodule (SPCG mean diameter, 16.2±5.1 mm). Mycobacteria, including Mycobacterium tuberculosis (11.8%), Mycobacterium avium (11.8%) Mycobacterium kansasii (23.5%) and other Mycobacterium spp. (5.9%), were isolated from 9 of the patients (52.9%). Concurrent lung cancer was present in 3 patients (17.6%). When microbial agents could not be isolated, the interferon-γ release assay was useful for diagnosis. Positron emission tomography was not found to be useful for differentiating SPCGs from lung cancer, or for differentiating tuberculomas from NTM pulmonary nodules (NTMPNs). NTMPNs in cases of SPCGs were diagnosed more frequently in men. The findings indicate that a course of observation may be sufficient for patients in whom an SPCG from NTM (NTMPN) is identified by VATS. However, the presence of concurrent lung cancer in certain cases indicates that malignancy should not necessarily be excluded, particularly in NTMPNs, and highlights the necessity of aggressive diagnosis by VATS.
Collapse
Affiliation(s)
- Hidehiro Watanabe
- Department of Infection Control and Prevention, Tokyo Medical University Hospital, Tokyo 160-0023, Japan
| | - Tomonori Uruma
- Department of Infection Control and Prevention, Tokyo Medical University Ibaraki Medical Center, Inashiki, Ibaraki 300-0395, Japan
| | - Ikuo Seita
- Department of Infection Control and Prevention, Tokyo Medical University Ibaraki Medical Center, Inashiki, Ibaraki 300-0395, Japan
| | - Tsuyoshi Oishi
- Department of Infection Control and Prevention, Tokyo Medical University Ibaraki Medical Center, Inashiki, Ibaraki 300-0395, Japan
| | - Yusuke Watanabe
- Department of Infection Control and Prevention, Tokyo Medical University Hospital, Tokyo 160-0023, Japan
| | - Ayaka Tsukimori
- Department of Infection Control and Prevention, Tokyo Medical University Hospital, Tokyo 160-0023, Japan
| | - Yoshiteru Haga
- Department of Infection Control and Prevention, Tokyo Medical University Hospital, Tokyo 160-0023, Japan
| | - Shinji Fukushima
- Department of Infection Control and Prevention, Tokyo Medical University Hospital, Tokyo 160-0023, Japan
| | - Akihiro Sato
- Department of Infection Control and Prevention, Tokyo Medical University Hospital, Tokyo 160-0023, Japan
| | - Itaru Nakamura
- Department of Infection Control and Prevention, Tokyo Medical University Hospital, Tokyo 160-0023, Japan
| | - Tetsuya Matsumoto
- Department of Microbiology, Tokyo Medical University Hospital, Tokyo 160-0023, Japan
| |
Collapse
|
28
|
Samir M, Glister C, Mattar D, Laird M, Knight PG. Follicular expression of pro-inflammatory cytokines tumour necrosis factor-α (TNFα), interleukin 6 (IL6) and their receptors in cattle: TNFα, IL6 and macrophages suppress thecal androgen production in vitro. Reproduction 2017; 154:35-49. [PMID: 28432091 DOI: 10.1530/rep-17-0053] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/06/2017] [Accepted: 04/21/2017] [Indexed: 12/21/2022]
Abstract
Pro-inflammatory cytokines secreted by macrophages and other cell types are implicated as intraovarian factors affecting different aspects of ovarian function including follicle and corpus luteum 'turnover', steroidogenesis and angiogenesis. Here, we compared granulosal (GC) and thecal (TC) expression of TNF, IL6 and their receptors (TNFRSF1A, TNFRSF1B and IL6R) during bovine antral follicle development; all five mRNA transcripts were detected in both GC and TC and statistically significant cell-type and follicle stage-related differences were evident. Since few studies have examined cytokine actions on TC steroidogenesis, we cultured TC under conditions that retain a non-luteinized 'follicular' phenotype and treated them with TNFα and IL6 under basal and LH-stimulated conditions. Both TNFα and IL6 suppressed androgen secretion concomitantly with CYP17A1 and LHCGR mRNA expression. In addition, TNFα reduced INSL3, HSD3B1 and NOS3 expression but increased NOS2 expression. IL6 also reduced LHCGR and STAR expression but did not affect HSD3B1, INSL3, NOS2 or NOS3 expression. As macrophages are a prominent source of these cytokines in vivo, we next co-cultured TC with macrophages and observed an abolition of LH-induced androgen production accompanied by a reduction in CYP17A1, INSL3, LHCGR, STAR, CYP11A1 and HSD3B1 expression. Exposure of TC to bacterial lipopolysaccharide also blocked LH-induced androgen secretion, an effect reduced by a toll-like receptor blocker (TAK242). Collectively, the results support an inhibitory action of macrophages on thecal androgen production, likely mediated by their secretion of pro-inflammatory cytokines that downregulate the expression of LHCGR, CYP17A1 and INSL3. Bovine theca interna cells can also detect and respond directly to lipopolysaccharide.
Collapse
Affiliation(s)
| | - Claire Glister
- School of Biological SciencesUniversity of Reading, Whiteknights, Reading, UK
| | - Dareen Mattar
- School of Biological SciencesUniversity of Reading, Whiteknights, Reading, UK
| | - Mhairi Laird
- School of Biological SciencesUniversity of Reading, Whiteknights, Reading, UK
| | - Phil G Knight
- School of Biological SciencesUniversity of Reading, Whiteknights, Reading, UK
| |
Collapse
|
29
|
Abstract
Males and females differ in their immunological responses to foreign and self-antigens and show distinctions in innate and adaptive immune responses. Certain immunological sex differences are present throughout life, whereas others are only apparent after puberty and before reproductive senescence, suggesting that both genes and hormones are involved. Furthermore, early environmental exposures influence the microbiome and have sex-dependent effects on immune function. Importantly, these sex-based immunological differences contribute to variations in the incidence of autoimmune diseases and malignancies, susceptibility to infectious diseases and responses to vaccines in males and females. Here, we discuss these differences and emphasize that sex is a biological variable that should be considered in immunological studies.
Collapse
|
30
|
Ghasemi M, Nematbakhsh M, Pezeshki Z, Soltani N, Moeini M, Talebi A. Nephroprotective effect of estrogen and progesterone combination on cisplatin-induced nephrotoxicity in ovariectomized female rats. Indian J Nephrol 2016; 26:167-75. [PMID: 27194830 PMCID: PMC4862261 DOI: 10.4103/0971-4065.160337] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Recently, we reported that estrogen (Es) has no beneficial effect on cisplatin (CP)-induced nephrotoxicity, but the role of progesterone (Pr) and the combination of Es and Pr are not yet well-defined. In this study, we investigated the protective role of Pr, and co-administration of Es/Pr on CP-induced nephrotoxicity. Eighty-six ovariectomized female Wistar rats were divided into 13 groups, and the experiments were performed in two phases. In Phase I, Groups 1-4 received 2, 5, 10, and 25 mg/kg, IM Pr dissolved in sesame oil every 5 days for four doses. Groups 5-8 had the same treatment regimen as Groups 1-4, but after the third injection the animals also received continuous dose of CP (2.5 mg/kg/day, i.p.) for 8 days. Group 9, as the positive control group, received sesame oil instead of Pr plus CP. Group 10, as the negative control group, received sesame oil instead of Pr. After the most effective dose of Pr was determined in Phase I, Groups 11-13 in Phase II received 10 mg/kg Pr plus either 0.25, 0.5, or 1 mg/kg, IM estradiol valerate every 5 days for four doses. After the third injection, they also received a continuous dose of CP for 8 days. The levels of blood urea nitrogen (BUN) and creatinine (Cr), kidney tissue damage score (KTDS), and kidney weight (KW) increased and body weight (BW) decreased in the positive control group (P < 0.05). Administration of Pr (10 mg/kg) plus CP decreased KTDS and BW loss and KW. Co-administration of ES/Pr at specific doses improved Cr, BUN, and KTDS; and resulted in reduced CP-induced nephrotoxicity. The results obtained suggest that the beneficial effect of Pr on CP-induced nephrotoxicity is dose-dependent. In addition, combination of Es/Pr with a specific dose decreased CP-induced nephrotoxicity.
Collapse
Affiliation(s)
- M Ghasemi
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - M Nematbakhsh
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran; Isfahan Institute of Basic and Applied Sciences Research, Isfahan, Iran
| | - Z Pezeshki
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - N Soltani
- Department of Physiology, Hormozgan University of Medical Sciences, Isfahan, Iran
| | - M Moeini
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - A Talebi
- Department of Clinical Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
31
|
Grimstad F, Krieg S. Immunogenetic contributions to recurrent pregnancy loss. J Assist Reprod Genet 2016; 33:833-47. [PMID: 27169601 DOI: 10.1007/s10815-016-0720-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022] Open
Abstract
While sporadic pregnancy loss is common, occurring in 15 % of pregnancies, recurrent pregnancy loss (RPL) impacts approximately 5 % of couples. Though multiple causes are known (including structural, hormonal, infectious, autoimmune, and thrombophilic causes), after evaluation, roughly half of all cases remain unexplained. The idiopathic RPL cases pose a challenging therapeutic dilemma in addition to incurring much physical and emotional morbidity. Immunogenetic causes have been postulated to contribute to these cases of RPL. Natural Killer cell, T cell expression pattern changes in the endometrium have both been shown in patients with RPL. Human leukocyte antigen (HLA) and cytokine allelic variations have also been studied as etiologies for RPL. Some of the results have been promising, however the studies are small and have not yet put forth outcomes that would change our current diagnosis and management of RPL. Larger database studies are needed with stricter control criteria before reasonable conclusions can be drawn.
Collapse
Affiliation(s)
- Frances Grimstad
- Department of Obstetrics and Gynecology, University of Kansas, 3901 Rainbow Blvd MS 2028, Kansas City, KS, 66160, USA.
| | - Sacha Krieg
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Oregon Health Sciences University, Portland, OR, USA
| |
Collapse
|
32
|
Salman K, Rashmi, Priti S, Molly M, Kumar VS, Zeenat S. Hepatitis B virus infection in pregnant women and transmission to newborns. ASIAN PACIFIC JOURNAL OF TROPICAL DISEASE 2015. [DOI: 10.1016/s2222-1808(15)60809-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
33
|
Flanagan KL. Sexual dimorphism in biomedical research: a call to analyse by sex. Trans R Soc Trop Med Hyg 2015; 108:385-7. [PMID: 24934286 DOI: 10.1093/trstmh/tru079] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Katie L Flanagan
- Department of Immunology, Monash University, Prahran, Melbourne, Victoria 3181, Australia
| |
Collapse
|
34
|
CCL2 and CCL5 Are Novel Therapeutic Targets for Estrogen-Dependent Breast Cancer. Clin Cancer Res 2015; 21:3794-805. [DOI: 10.1158/1078-0432.ccr-15-0204] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/09/2015] [Indexed: 11/16/2022]
|
35
|
Lu J, Reese J, Zhou Y, Hirsch E. Progesterone-induced activation of membrane-bound progesterone receptors in murine macrophage cells. J Endocrinol 2015; 224:183-94. [PMID: 25472814 PMCID: PMC4297269 DOI: 10.1530/joe-14-0470] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Parturition is an inflammatory process mediated to a significant extent by macrophages. Progesterone (P4) maintains uterine quiescence in pregnancy, and a proposed functional withdrawal of P4 classically regulated by nuclear progesterone receptors (nPRs) leads to labor. P4 can affect the functions of macrophages despite the reported lack of expression of nPRs in these immune cells. Therefore, in this study we investigated the effects of the activation of the putative membrane-associated PR on the function of macrophages (a key cell for parturition) and discuss the implications of these findings for pregnancy and parturition. In murine macrophage cells (RAW 264.7), activation of mPRs by P4 modified to be active only extracellularly by conjugation to BSA (P4BSA, 1.0×10(-7) mol/l) caused a pro-inflammatory shift in the mRNA expression profile, with significant upregulation of the expression of cyclooxygenase 2 (COX2 (Ptgs2)), Il1B, and Tnf and downregulation of membrane progesterone receptor alpha (Paqr7) and oxytocin receptor (Oxtr). Pretreatment with PD98059, a MEK1/2 inhibitor, significantly reduced P4BSA-induced expression of mRNA of Il1B, Tnf, and Ptgs2. Inhibition of protein kinase A (PKA) by H89 blocked P4BSA-induced expression of Il1B and Tnf mRNA. P4BSA induced rapid phosphorylation of MEK1/2 and CREB (a downstream target of PKA). This phosphorylation was inhibited by pretreatment with PD98059 and H89, respectively, revealing that MEK1/2 and PKA are two of the components involved in mPR signaling. Taken together, these results indicate that changes in membrane progesterone receptor alpha expression and signaling in macrophages are associated with the inflammatory responses; and that these changes might contribute to the functional withdrawal of P4 related to labor.
Collapse
Affiliation(s)
- Jing Lu
- Department of OB/GYNNorthShore University HealthSystem, 2650 Ridge Avenue, Evanston, Illinois 60201, USADepartment of OB/GYNPritzker School of Medicine, University of Chicago, 924 East 57th Street Suite 104, Chicago, Illinois 60637, USA Department of OB/GYNNorthShore University HealthSystem, 2650 Ridge Avenue, Evanston, Illinois 60201, USADepartment of OB/GYNPritzker School of Medicine, University of Chicago, 924 East 57th Street Suite 104, Chicago, Illinois 60637, USA
| | - Joshua Reese
- Department of OB/GYNNorthShore University HealthSystem, 2650 Ridge Avenue, Evanston, Illinois 60201, USADepartment of OB/GYNPritzker School of Medicine, University of Chicago, 924 East 57th Street Suite 104, Chicago, Illinois 60637, USA
| | - Ying Zhou
- Department of OB/GYNNorthShore University HealthSystem, 2650 Ridge Avenue, Evanston, Illinois 60201, USADepartment of OB/GYNPritzker School of Medicine, University of Chicago, 924 East 57th Street Suite 104, Chicago, Illinois 60637, USA
| | - Emmet Hirsch
- Department of OB/GYNNorthShore University HealthSystem, 2650 Ridge Avenue, Evanston, Illinois 60201, USADepartment of OB/GYNPritzker School of Medicine, University of Chicago, 924 East 57th Street Suite 104, Chicago, Illinois 60637, USA Department of OB/GYNNorthShore University HealthSystem, 2650 Ridge Avenue, Evanston, Illinois 60201, USADepartment of OB/GYNPritzker School of Medicine, University of Chicago, 924 East 57th Street Suite 104, Chicago, Illinois 60637, USA
| |
Collapse
|
36
|
Avivi I, Farbstein D, Brenner B, Horowitz NA. Non-Hodgkin lymphomas in pregnancy: tackling therapeutic quandaries. Blood Rev 2014; 28:213-20. [PMID: 25108745 DOI: 10.1016/j.blre.2014.06.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 06/23/2014] [Accepted: 06/27/2014] [Indexed: 12/15/2022]
Abstract
Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL) often present with systemic symptoms such as fatigue, shortness of breath and night sweats, mimicking pregnancy-related features which may result in delayed disease diagnosis. Furthermore, the wish to avoid investigational imaging, aiming to protect the fetus from radiation exposure, may lead to a further delay, which does not often result in significant changes in HL clinical nature and patient outcome. In contrast, a more aggressive behavior (i.e., advanced disease stage and reproductive organ involvement) of most NHL types diagnosed in pregnancy may require urgent therapeutic intervention to prevent disease progression. Current management of pregnancy-associated NHL depends on histological subtype of the disease, gestational stage at diagnosis and the urgency of treatment for a specific patient. Patients diagnosed with indolent lymphoma may often be just followed, whereas those presenting with aggressive or highly aggressive disease need to be urgently treated with chemoimmunotherapy, either after undergoing an elective pregnancy termination if diagnosed at an early gestational stage, or with pregnancy preservation, if diagnosed later. Supportive care of NHL is also important; however, granulocyte colony stimulating factor (G-CSF) which is commonly used outside of pregnancy, should be cautiously employed, considering its established teratogenicity in animals, though this is less proven in humans. In conclusion, given the paucity of studies prospectively evaluating the outcome of pregnant women with NHL, international efforts are warranted to elucidate critical issues and develop guidelines for the management of such patients.
Collapse
Affiliation(s)
- Irit Avivi
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel; Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| | - Dan Farbstein
- Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Benjamin Brenner
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel; Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Netanel A Horowitz
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel; Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
37
|
Hamlin HJ, Lowers RH, Kohno S, Mitsui-Watanabe N, Amano H, Hara A, Ohta Y, Miyagawa S, Iguchi T, Guillette LJ. The reproductive hormone cycle of adult female American alligators from a barrier island population. Reproduction 2014; 147:855-63. [DOI: 10.1530/rep-14-0031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Comparatively, little data are available detailing the geographic variation that exists in the reproductive endocrinology of adult alligators, especially those living in barrier islands. The Merritt Island National Wildlife Refuge (MI) is a unique barrier island environment and home to the Kennedy Space Center (FL, USA). Seasonal patterns of sex steroids were assessed in adult female American alligators from MI monthly from 2008 to 2009, with additional samples collected at more random intervals in 2006, 2007, and 2010. Plasma 17β-estradiol and vitellogenin concentrations peaked in April, coincident with courtship and mating, and showed patterns similar to those observed in adult female alligators in other regions. Plasma concentrations of progesterone, however, showed patterns distinctly different than those reported for alligator populations in other regions and remained relatively constant throughout the year. Plasma DHEA peaked in July around the time of oviposition, decreased in August, and then remained constant for the remaining months, except for a moderate increase in October. Circulating concentrations of DHEA have not been previously assessed in a female crocodilian, and plasma concentrations coincident with reproductive activity suggest a reproductive and/or behavioral role. Interestingly, plasma testosterone concentrations peaked in May of 2008, as has been shown in female alligator populations in other regions, but showed no peak in 2009, demonstrating dramatic variability from year to year. Surveys showed 2009 to be particularly depauperate of alligator nests in MI, and it is possible that testosterone could serve as a strong indicator of breeding success.
Collapse
|
38
|
Hassouna A, Obaia E, Marzouk S, Rateb M, Haidara M. The role of sex hormones in induced-systemic inflammation in female albino rats. ACTA PHYSIOLOGICA HUNGARICA 2014; 101:112-127. [PMID: 24631798 DOI: 10.1556/aphysiol.101.2014.1.12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
UNLABELLED Estrogen (E(2)) and progesterone (P) hormones have a pro-inflammatory and an anti-inflammatory role under different conditions. The current study explored this phenomenon in the context of septic inflammation. MATERIALS AND METHODS This study involved 48 female albino rats. E(2) (4 mg/100 g body weight (b.w.) and P (5 mg/kg b.w.) were administered to ovariectomized (OVX) rats after systemic inflammation (SI) induced by puncturing the caecum I cm from its end with a single hole by using a 21-gauge needle. Key indices of inflammation and apoptosis were evaluated. RESULTS OVX animals subjected to SI showed significantly increased levels of serum tumor necrosis factor-alpha (TNF-u), C reactive protein (CRP) and alanine aminotransferase (ALT). They also showed higher levels of expression of the enzyme inducible nitric oxide synthase (iN OS); 312 ± 43 mg/ml; in the liver, and the activity of both cyclooxygenase 2 (COX-2); 59.4 ± 3.2 U/ml; and caspase 3 enzymes; 6.3 ± 0.54 ng/ml; when compared to non-OVX animals subjected to (SI), (180 ± 3 mg/ml, 16.4 ± 1.69 U/ml, 0.98 ± 0.23 ng/ml respectively). Administration of E(2) resulted in a significant reduction of all serum and liver tissue parameters of inflammation (e.g.decreased iNOS; 193 ± 28 mg/ml and COX-2; 27.6 ± 3.91 U/ml) and decreased apoptosis (Caspase 3; 1.18 ± 0.21 ng/ml). In contrast, OVX animals injected with P before induction of SI showed a significant rise of all measured parameters. CONCLUSIONS E(2) and Pin physiological levels have contrasting though complementary roles in regulation of the immune system possibly allowing a limited inflammatory response while preventing excessive damage to the tissues.
Collapse
Affiliation(s)
- A Hassouna
- Cairo University Departments of Medical Biochemistry, Faculty of Medicine Cairo Egypt
| | - E Obaia
- Cairo University Departments of Medical Biochemistry, Faculty of Medicine Cairo Egypt
| | - S Marzouk
- Cairo University Departments of Medical Biochemistry, Faculty of Medicine Cairo Egypt
| | - M Rateb
- Cairo University Departments of Physiology, Faculty of Medicine Cairo Egypt
| | - Mohamed Haidara
- King Khalid University Department of Physiology, College of Medicine Abha Saudi Arabia
| |
Collapse
|
39
|
Do androgen deprivation drugs affect the immune cross-talk between mononuclear and prostate cancer cells? Biomed Pharmacother 2013; 68:21-4. [PMID: 24406295 DOI: 10.1016/j.biopha.2013.12.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/10/2013] [Indexed: 12/13/2022] Open
Abstract
The aim of the study was to examine the effect of androgen deprivation drugs, i.e. leuprolide and bicalutamide on the immune cross-talk between human peripheral blood mononuclear cells (PBMC) and cells from PC-3 and LNCaP human prostate cancer lines. PBMC, PC-3 and LNCaP were separately incubated without and with two androgen-deprivation drugs, i.e. leuprolide and bicalutamide, and the secretion of IL-1β, IL-6, IL-1ra and IL-10 was examined. In addition, the effect of both drugs on the production of those cytokines was carried out after 24 hours incubation of PBMC with both types of cancer cells. Leuprolide or bicalutamide did not affect the production of the cytokines by PBMC or by the prostate cancer cells from the two lines. Incubation of PBMC with PC-3 or LNCaP cells caused increased production of IL-1β, IL-6 and IL-10 as compared with PBMC incubated without malignant cells. While 10(-7) M and 10(-8) M of leuprolide caused a decreased secretion of IL-1β by PBMC previously incubated with prostate cancer cells without the drug, bicalutamide did not affect this PBMC activity at any drug concentration. This observation suggests the existence of an additional mechanism explaining the effect of androgen deprivation therapy in prostate cancer patients.
Collapse
|
40
|
Sayama S, Nagamatsu T, Schust DJ, Itaoka N, Ichikawa M, Kawana K, Yamashita T, Kozuma S, Fujii T. Human decidual macrophages suppress IFN-γ production by T cells through costimulatory B7-H1:PD-1 signaling in early pregnancy. J Reprod Immunol 2013; 100:109-17. [DOI: 10.1016/j.jri.2013.08.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 07/29/2013] [Accepted: 08/02/2013] [Indexed: 12/12/2022]
|
41
|
Flores-Espinosa P, Pineda-Torres M, Vega-Sánchez R, Estrada-Gutiérrez G, Espejel-Nuñez A, Flores-Pliego A, Maida-Claros R, Paredes-Vivas Y, Morales-Méndez I, Sosa-González I, Chávez-Mendoza A, Zaga-Clavellina V. Progesterone elicits an inhibitory effect upon LPS-induced innate immune response in pre-labor human amniotic epithelium. Am J Reprod Immunol 2013; 71:61-72. [PMID: 24128422 DOI: 10.1111/aji.12163] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 09/09/2013] [Indexed: 12/19/2022] Open
Abstract
PROBLEM Infection of human fetal membranes elicits secretion of pro-inflammatory modulators through its innate immune capacities. We investigated the effect of lipopolysacharide (LPS) and progesterone (P4) upon expression of TLR-4/MyD88, TNFα, IL-6, IL-8, IL-10, and HBD2 on the human amniotic epithelium. METHOD OF STUDY Explants of the human amniotic epithelium were pre-treated with 0.01, 0.1, and 1.0 μM of P4; then cotreated with 1000 ng/mL LPS. TLR-4 was immuno-detected, and concentrations of MyD88, TNFα, IL-6, IL-8, IL-10, and HBD2 were quantified by ELISA. RESULTS P4 significantly reduced the expression of LPS-induced TLR-4/MyD88. LPS increased the concentrations of TNFα, IL-6, IL-8, IL-10, and HBD2 by factors of 30-, eight, three, three, and fivefold, respectively. P4 at 1.0 μM was the most effective dose to blunt the secretion of TNFα, IL-6, and HBD-2. RU-486 blocks the effect of P4. CONCLUSION P4 inhibited LPS-induced TLR-4/MyD88 and pro-inflammatory factors in the human amniotic epithelium. These results could explain partially how P4 can protect the amniotic region of fetal membranes and generate a compensatory mechanism that limits the secretion of pro-inflammatory modulators, which could jeopardize the immune privilege during pregnancy.
Collapse
Affiliation(s)
- Pilar Flores-Espinosa
- Department of Cell Biology, Instituto Nacional de Perinatología 'Isidro Espinosa de los Reyes', Mexico City, Mexico; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Reproductive organ involvement in non-Hodgkin lymphoma during pregnancy: a systematic review. Lancet Oncol 2013; 14:e275-82. [DOI: 10.1016/s1470-2045(12)70589-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
43
|
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: 124] [Impact Index Per Article: 9.5] [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
|
44
|
Robinson DP, Klein SL. Pregnancy and pregnancy-associated hormones alter immune responses and disease pathogenesis. Horm Behav 2012; 62:263-71. [PMID: 22406114 PMCID: PMC3376705 DOI: 10.1016/j.yhbeh.2012.02.023] [Citation(s) in RCA: 461] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 02/20/2012] [Accepted: 02/22/2012] [Indexed: 12/12/2022]
Abstract
During pregnancy, it is evolutionarily advantageous for inflammatory immune responses that might lead to fetal rejection to be reduced and anti-inflammatory responses that promote transfer of maternal antibodies to the fetus to be increased. Hormones modulate the immunological shift that occurs during pregnancy. Estrogens, including estradiol and estriol, progesterone, and glucocorticoids increase over the course of pregnancy and affect transcriptional signaling of inflammatory immune responses at the maternal-fetal interface and systemically. During pregnancy, the reduced activity of natural killer cells, inflammatory macrophages, and helper T cell type 1 (Th1) cells and production of inflammatory cytokines, combined with the higher activity of regulatory T cells and production of anti-inflammatory cytokines, affects disease pathogenesis. The severity of diseases caused by inflammatory responses (e.g., multiple sclerosis) is reduced and the severity of diseases that are mitigated by inflammatory responses (e.g., influenza and malaria) is increased during pregnancy. For some infectious diseases, elevated inflammatory responses that are necessary to control and clear a pathogen have a negative consequence on the outcome of pregnancy. The bidirectional interactions between hormones and the immune system contribute to both the outcome of pregnancy and female susceptibility to disease.
Collapse
Affiliation(s)
- Dionne P. Robinson
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205
| | - Sabra L. Klein
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205
| |
Collapse
|
45
|
Quintar AA, Leimgruber C, Pessah OA, Doll A, Maldonado CA. Androgen depletion augments antibacterial prostate host defences in rats. ACTA ACUST UNITED AC 2012; 35:845-859. [DOI: 10.1111/j.1365-2605.2012.01288.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
46
|
Macrophage secretions modulate the steroidogenesis of polycystic ovary in rats: effect of testosterone on macrophage pro-inflammatory cytokines. Life Sci 2012; 90:733-9. [PMID: 22480516 DOI: 10.1016/j.lfs.2012.03.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 02/16/2012] [Accepted: 03/16/2012] [Indexed: 11/23/2022]
Abstract
AIMS The macrophage secretions' effect on ovarian steroidogenesis is investigated in a polycystic ovary syndrome rat model (PCO rat). The influence of testosterone environment on the expression of macrophage pro-inflammatory cytokines that participate in ovarian steroidogenesis is studied. MAIN METHODS PCO rats were induced by estradiol valerate. Spleen macrophages were cultured with and without testosterone (10(-6) M) and their secretions were used to stimulate ovaries from PCO and control rats. Ovarian hormones released and ovary mRNA levels of P450 aromatase and 3β-hydroxysteroid dehydrogenase were measured by radioimmunoassay and RT-PCR, respectively. The tumor necrosis factor alpha (TNFα) and nitric oxide (NO) levels in macrophage culture medium, along with the TNFα, interleukin (IL)-6, IL-10 and androgen receptors (AR) mRNA levels in macrophage cells were determined. KEY FINDINGS Macrophages from PCO rats released more TNFα and NO, expressed higher TNFα and IL-6, lower AR, and no change in IL-10 mRNA levels than control macrophages. TNFα, IL-6 and AR changes were greater after macrophage testosterone treatment. Macrophage secretions from PCO rats stimulated androstenedione and decreased estradiol release and ovarian mRNA P450 aromatase expression in PCO rats compared to macrophage secretions from control rats. These effects were greater when macrophages from PCO rats were treated with testosterone. Ovarian progesterone response was unchanged. SIGNIFICANCE The differential steroidogenic ability of macrophage secretions from PCO rats is associated to the in vitro testosterone environment. Testosterone, probably acting on macrophage AR, induces a greater release of TNFα, modifying ovarian response by increasing androstenedione and slightly decreasing estradiol without affecting progesterone.
Collapse
|
47
|
Menzies FM, Henriquez FL, Alexander J, Roberts CW. Selective inhibition and augmentation of alternative macrophage activation by progesterone. Immunology 2011; 134:281-91. [PMID: 21977998 DOI: 10.1111/j.1365-2567.2011.03488.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Progesterone is the female sex hormone necessary for the maintenance of pregnancy, and is known to modulate macrophage activation. However, studies have concentrated exclusively on the ability of progesterone to negatively regulate the innate and classical pathways of activation, associated with nitric oxide (NO) and interleukin (IL)-12 production. Our aim was to examine the ability of progesterone to modulate alternative macrophage activation. Bone marrow cells were isolated and differentiated from male BALB/c mice, exposed to varying concentrations of progesterone and stimulated with lipopolysaccharide (LPS) (innate activation), IL-4 (alternative activation) or LPS in combination with IL-4. Our present study demonstrates that progesterone not only down-regulates inducible nitric oxide synthase 2 (iNOS) activity in macrophages but also arginase activity, in a dose-dependent manner, independent of the stimuli, whether it is induced by LPS (innate activation), IL-4 (alternative activation) or LPS in combination with IL-4. The ability of progesterone to down-modulate IL-4-induced cell surface expression of the mannose receptor further suggested a negative regulation of alternative macrophage activation by this hormone. Analysis of mRNA expression, by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), of genes associated with innate and alternative macrophage activation revealed that progesterone down-regulated LPS-induced macrophage nos2, argI and p40 (IL-12/IL-23) expression and IL-4-induced argI, mrc-1 and fizz1 expression. However, progesterone up-regulated IL-4-induced macrophage expression of ym1, while dectin-1 expression remained unaltered. Following treatment of macrophages with LPS and IL-4 in combination a similar pattern was observed, with the exception that progesterone up-regulated macrophage expression of fizz1 as well as ym1 and did not modify mrc-1 expression. Our data demonstrate for the first time that a hormone has the ability to regulate selectively the expression of different genes associated with alternative macrophage activation.
Collapse
Affiliation(s)
- Fiona M Menzies
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | | | | | | |
Collapse
|
48
|
Fu Y, Li L, Liu X, Ma C, Zhang J, Jiao Y, You L, Chen ZJ, Zhao Y. Estrogen promotes B cell activation in vitro through down-regulating CD80 molecule expression. Gynecol Endocrinol 2011; 27:593-6. [PMID: 21726119 DOI: 10.3109/09513590.2010.507281] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Estrogen is the main female hormone of women. It has diverse effects on cell growth, differentiation and homeostatic functions. Accumulated evidence has indicated that estrogen may regulate multiple immune functions and the immune status of women. However, there is little report on the effect of estrogen on mature B cell functions. In this study, we observed the effect of 17β-estradiol (E2) on the proliferation, apoptosis, antibody production and differentiation of splenic B cells of mice in vitro. Splenocytes of female BALB/c mice were isolated and cultured with E2. E2 treatments decreased the expression of CD80 molecule on splenic B cells but enhanced the total IgG antibody production of splenocyte, without promoting the differentiation of B cells to plasma cells. E2 protected splenic B cells from the serum-deficiency-induced apoptosis but had no influence on the proliferation of B cells. These results suggest that estrogen may promote the activity of B cells through down-regulating the expression of CD80 molecule on B cells.
Collapse
Affiliation(s)
- Yibing Fu
- Key Laboratory for Reproductive Medicine of Shandong Province, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Robert R, Ghazali DA, Favreau F, Mauco G, Hauet T, Goujon JM. Gender difference and sex hormone production in rodent renal ischemia reperfusion injury and repair. JOURNAL OF INFLAMMATION-LONDON 2011; 8:14. [PMID: 21658244 PMCID: PMC3127739 DOI: 10.1186/1476-9255-8-14] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 06/09/2011] [Indexed: 01/29/2023]
Abstract
BACKGROUND Several lines of evidence suggest a protective effect of female sex hormones in several organs subjected to ischemia-reperfusion injury. The aim of the study was to investigate sex hormone production in male rats after a renal ischemia-reperfusion sequence and analyze the influence of gender differences on tissue remodelling during the recovery process. METHOD Age-matched sexually mature male and female rats were subjected to 60 min of renal unilateral ischemia by pedicle clamping with contralateral nephrectomy and followed for 1 or 5 days after reperfusion. Plasma creatinine, systemic testosterone, progesterone and estradiol levels were determined. Tubular injury, cell proliferation and inflammation, were evaluated as well as proliferating cell nuclear antigen, vimentin and translocator protein (TSPO) expressions by immunohistochemistry. RESULTS After 1 and 5 days of reperfusion, plasma creatinine was significantly higher in males than in females, supporting the high mortality in this group. After reperfusion, plasma testosterone levels decreased whereas estradiol significantly increased in male rats. Alterations of renal function, associated with tubular injury and inflammation persisted during the 5 days post-ischemia-reperfusion, and a significant improvement was observed in females at 5 days of reperfusion. Proliferating cell nuclear antigen and vimentin expression were upregulated in kidneys from males and attenuated in females, in parallel to injury development. TSPO expression was transiently increased in proximal tubules in male rats. CONCLUSIONS After ischemia, renal function recovery and tissue injury is gender-dependent. These differences are associated with a modulation of sex hormone production and a modification of tissue remodeling and proliferative cell processes.
Collapse
Affiliation(s)
- René Robert
- CHU Poitiers, Service de Réanimation Médicale Poitiers, F-86000, France.
| | | | | | | | | | | |
Collapse
|
50
|
Larcombe AN, Foong RE, Bozanich EM, Berry LJ, Garratt LW, Gualano RC, Jones JE, Dousha LF, Zosky GR, Sly PD. Sexual dimorphism in lung function responses to acute influenza A infection. Influenza Other Respir Viruses 2011; 5:334-42. [PMID: 21668688 PMCID: PMC4942045 DOI: 10.1111/j.1750-2659.2011.00236.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Please cite this paper as: Larcombe et al. (2011) Sexual dimorphism in lung function responses to acute influenza A infection. Influenza and Other Respiratory Viruses 5(5), 334–342. Background Males are generally more susceptible to respiratory infections; however, there are few data on the physiological responses to such infections in males and females. Objectives To determine whether sexual dimorphism exists in the physiological/inflammatory responses of weanling and adult BALB/c mice to influenza. Methods Weanling and adult mice of both sexes were inoculated with influenza A or appropriate control solution. Respiratory mechanics, responsiveness to methacholine (MCh), viral titre and bronchoalveolar lavage (BAL) cellular inflammation/cytokines were measured 4 (acute) and 21 (resolution) days post‐inoculation. Results Acute infection impaired lung function and induced hyperresponsiveness and cellular inflammation in both sexes at both ages. Males and females responded differently with female mice developing greater abnormalities in tissue damping and elastance and greater MCh responsiveness at both ages. BAL inflammation, cytokines and lung viral titres were similar between the sexes. At resolution, all parameters had returned to baseline levels in adults and weanling males; however, female weanlings had persisting hyperresponsiveness. Conclusions We identified significant differences in the physiological responses of male and female mice to infection with influenza A, which occurred in the absence of variation in viral titre and cellular inflammation.
Collapse
Affiliation(s)
- Alexander N Larcombe
- Division of Clinical Sciences, Telethon Institute for Child Health Research, Centre for Child Health Research, University of Western Australia, West Perth, WA, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|