1
|
Yoon C, Kim HK, Ham YS, Gil WJ, Mun SJ, Cho E, Yuk JM, Yang CS. Toxoplasma gondii macrophage migration inhibitory factor shows anti- Mycobacterium tuberculosis potential via AZIN1/STAT1 interaction. SCIENCE ADVANCES 2024; 10:eadq0101. [PMID: 39453997 PMCID: PMC11506136 DOI: 10.1126/sciadv.adq0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/20/2024] [Indexed: 10/27/2024]
Abstract
Mycobacterium tuberculosis (MTB) is a pathogenic bacterium, belonging to the family Mycobacteriaceae, that causes tuberculosis (TB). Toxoplasma gondii macrophage migration inhibitory factor (TgMIF), a protein homolog of macrophage migration inhibitory factor, has been explored for its potential to modulate immune responses during MTB infections. We observed that TgMIF that interacts with CD74, antizyme inhibitor 1 (AZIN1), and signal transducer and activator of transcription 1 (STAT1) modulates endocytosis, restoration of mitochondrial function, and macrophage polarization, respectively. These interactions promote therapeutic efficacy in mice infected with MTB, thereby presenting a potential route to host-directed therapy development. Furthermore, TgMIF, in combination with first-line TB drugs, significantly inhibited drug-resistant MTB strains, including multidrug-resistant TB. These results demonstrate that TgMIF is potentially a multifaceted therapeutic agent against TB, acting through immune modulation, enhancement of mitochondrial function, and dependent on STAT1 and AZIN1 pathways.
Collapse
Affiliation(s)
- Chanjin Yoon
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
- Institute of Natural Science & Technology, Hanyang University, Ansan 15588, South Korea
| | - Hyo Keun Kim
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
| | - Yu Seong Ham
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
| | - Woo Jin Gil
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
| | - Seok-Jun Mun
- Department of Bionano Engineering, Hanyang University, Seoul 04673, South Korea
| | - Euni Cho
- Department of Bionano Engineering, Hanyang University, Seoul 04673, South Korea
| | - Jae-Min Yuk
- Department of Infection Biology and Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Chul-Su Yang
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
- Department of Medicinal and Life Science, Hanyang University, Ansan 15588, South Korea
| |
Collapse
|
2
|
Yagasaki Y, Katayama Y, Kinoshita Y, Nagata T, Kawakami Y, Miyata M. Macrophages are activated in the rat anterior pituitary under chronic inflammatory conditions. Neurosci Lett 2021; 748:135688. [PMID: 33548409 DOI: 10.1016/j.neulet.2021.135688] [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: 06/27/2020] [Revised: 12/27/2020] [Accepted: 01/26/2021] [Indexed: 11/28/2022]
Abstract
In the anterior lobe of the pituitary gland (AP), non-endocrine cells regulate hormone secretion by endocrine cells. However, the functions of non-endocrine cells in the AP during chronic pain are largely unclear. Here, we show that macrophages, but not folliculostellate (FS) cells, were selectively increased in the AP in the complete Freund's adjuvant (CFA)-induced chronic inflammatory pain model in rats. In addition, IL-1β expression was increased in the AP, and the IL-1β-immunopositive cells were identified as macrophages. On the other hand, increased macrophage density and IL-1β expression were not detected in a neuropathic pain model induced by partial sciatic nerve ligation (PSL). Furthermore, we found c-Fos expression specifically in the somatotrophs under the chronic inflammatory pain condition. Because IL-1β promotes growth hormone (GH) synthesis and release, our results suggest that AP macrophage contributes to GH release through IL-1βduring chronic inflammatory pain. .
Collapse
Affiliation(s)
- Yuki Yagasaki
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Yoko Katayama
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Yoko Kinoshita
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan; Department of Anesthesiology, International University of Health and Welfare, Mita Hospital, Japan
| | - Tomonari Nagata
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Yoriko Kawakami
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Mariko Miyata
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan.
| |
Collapse
|
3
|
Brakel K, Hook MA. SCI and depression: Does inflammation commandeer the brain? Exp Neurol 2019; 320:112977. [PMID: 31203113 DOI: 10.1016/j.expneurol.2019.112977] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/29/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022]
Abstract
The incidence of depression is almost twice as high in the spinally injured population compared to the general population. While this incidence has long been attributed to the psychological, economic, and social burdens that accompany spinal cord injury (SCI), data from animal studies indicate that the biology of SCI may play an important role in the development of depression. Inflammation has been shown to impact stress response in rodents and humans, and inflammatory cytokines have been associated with depression for decades. The inflammation inherent to SCI may disrupt necessary mechanisms of mental homeostasis, such as serotonin production, dopamine production, and the hypothalamic pituitary adrenal axis. Additionally, gut dysbiosis that occurs after SCI can exacerbate inflammation and may cause further mood and behavior changes. These mediators combined may significantly contribute to the rise in depression seen after SCI. Currently, there are no therapies specific to depression after SCI. Elucidation of the molecular pathways that contribute to SCI-specific depression is crucial for the understanding of this disease and its potential treatments.
Collapse
Affiliation(s)
- Kiralyn Brakel
- School of Medicine, Department of Neuroscience and Experimental Therapeutics, Texas A&M University, Medical Research and Education Building, Ste. 1005, 8447 Riverside Pkwy, Bryan, TX 77807, United States; Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474 College Station, TAMU, TX, United States.
| | - Michelle A Hook
- School of Medicine, Department of Neuroscience and Experimental Therapeutics, Texas A&M University, Medical Research and Education Building, Ste. 1005, 8447 Riverside Pkwy, Bryan, TX 77807, United States; Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474 College Station, TAMU, TX, United States
| |
Collapse
|
4
|
Yelamanchi SD, Tyagi A, Mohanty V, Dutta P, Korbonits M, Chavan S, Advani J, Madugundu AK, Dey G, Datta KK, Rajyalakshmi M, Sahasrabuddhe NA, Chaturvedi A, Kumar A, Das AA, Ghosh D, Jogdand GM, Nair HH, Saini K, Panchal M, Sarvaiya MA, Mohanraj SS, Sengupta N, Saxena P, Subramani PA, Kumar P, Akkali R, Reshma SV, Santhosh RS, Rastogi S, Kumar S, Ghosh SK, Irlapati VK, Srinivasan A, Radotra BD, Mathur PP, Wong GW, Satishchandra P, Chatterjee A, Gowda H, Bhansali A, Pandey A, Shankar SK, Mahadevan A, Prasad TSK. Proteomic Analysis of the Human Anterior Pituitary Gland. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 22:759-769. [PMID: 30571610 DOI: 10.1089/omi.2018.0160] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The pituitary function is regulated by a complex system involving the hypothalamus and biological networks within the pituitary. Although the hormones secreted from the pituitary have been well studied, comprehensive analyses of the pituitary proteome are limited. Pituitary proteomics is a field of postgenomic research that is crucial to understand human health and pituitary diseases. In this context, we report here a systematic proteomic profiling of human anterior pituitary gland (adenohypophysis) using high-resolution Fourier transform mass spectrometry. A total of 2164 proteins were identified in this study, of which 105 proteins were identified for the first time compared with high-throughput proteomic-based studies from human pituitary glands. In addition, we identified 480 proteins with secretory potential and 187 N-terminally acetylated proteins. These are the first region-specific data that could serve as a vital resource for further investigations on the physiological role of the human anterior pituitary glands and the proteins secreted by them. We anticipate that the identification of previously unknown proteins in the present study will accelerate biomedical research to decipher their role in functioning of the human anterior pituitary gland and associated human diseases.
Collapse
Affiliation(s)
| | - Ankur Tyagi
- 2 Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Varshasnata Mohanty
- 2 Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Pinaki Dutta
- 3 Department of Endocrinology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Márta Korbonits
- 4 Department of Endocrinology, Barts and the London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Sandip Chavan
- 1 Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - Jayshree Advani
- 1 Institute of Bioinformatics, International Technology Park, Bangalore, India.,5 Manipal Academy of Higher Education, Manipal, India
| | - Anil K Madugundu
- 1 Institute of Bioinformatics, International Technology Park, Bangalore, India.,5 Manipal Academy of Higher Education, Manipal, India.,6 Center for Molecular Medicine, National Institute of Mental Health & Neurosciences, Bangalore, India.,7 Department of Laboratory Medicine and Pathology and Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| | - Gourav Dey
- 1 Institute of Bioinformatics, International Technology Park, Bangalore, India.,5 Manipal Academy of Higher Education, Manipal, India
| | - Keshava K Datta
- 1 Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - M Rajyalakshmi
- 8 Department of Biotechnology, BMS College of Engineering, Bangalore, India
| | | | - Abhishek Chaturvedi
- 9 Department of Biochemistry, Melaka Manipal Medical College, Manipal Academy of Higher Education, Manipal, India
| | - Amit Kumar
- 10 Institute of Life Sciences, Nalco Square, Bhubaneswar, India
| | - Apabrita Ayan Das
- 11 Cell Biology and Physiology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Dhiman Ghosh
- 12 Protein Engineering and Neurobiology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, India
| | | | - Haritha H Nair
- 13 Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Keshav Saini
- 14 Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | - Manoj Panchal
- 15 Department of Life Science, Central University of South Bihar, Gaya, India
| | | | - Soundappan S Mohanraj
- 17 Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Nabonita Sengupta
- 18 Neuroinflammation Laboratory, National Brain Research Centre, Manesar, India
| | - Priti Saxena
- 14 Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | | | - Pradeep Kumar
- 20 Department of Biotechnology, VBS Purvanchal University, Jaunpur, India
| | - Rakhil Akkali
- 21 Department of Biotechnology, Indian Institute of Technology, Madras, India
| | | | | | - Sangita Rastogi
- 24 Microbiology Laboratory, National Institute of Pathology, New Delhi, India
| | - Sudarshan Kumar
- 25 Proteomics and Structural Biology Laboratory, Animal Biotechnology Center, National Dairy Research Institute, Karnal, India
| | - Susanta Kumar Ghosh
- 19 Department of Molecular Parasitology, National Institute of Malaria Research, Bangalore, India
| | | | - Anand Srinivasan
- 27 Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bishan Das Radotra
- 28 Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Premendu P Mathur
- 29 Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Pondicherry, India
| | - G William Wong
- 30 Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Aditi Chatterjee
- 1 Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - Harsha Gowda
- 1 Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - Anil Bhansali
- 3 Department of Endocrinology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Akhilesh Pandey
- 1 Institute of Bioinformatics, International Technology Park, Bangalore, India.,5 Manipal Academy of Higher Education, Manipal, India.,6 Center for Molecular Medicine, National Institute of Mental Health & Neurosciences, Bangalore, India.,7 Department of Laboratory Medicine and Pathology and Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota.,32 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,33 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland.,34 Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,35 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Susarla K Shankar
- 36 Department of Neuropathology, National Institute of Mental Health and Neuro Sciences, Bangalore, India.,37 Human Brain Tissue Repository, National Institute of Mental Health and Neuro Sciences, Neurobiology Research Centre, Bangalore, India
| | - Anita Mahadevan
- 36 Department of Neuropathology, National Institute of Mental Health and Neuro Sciences, Bangalore, India.,37 Human Brain Tissue Repository, National Institute of Mental Health and Neuro Sciences, Neurobiology Research Centre, Bangalore, India
| | - T S Keshava Prasad
- 1 Institute of Bioinformatics, International Technology Park, Bangalore, India.,2 Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| |
Collapse
|
5
|
Lipschutz R, Bick J, Nguyen V, Lee M, Leng L, Grigorenko E, Bucala R, Mayes LC, Crowley MJ. Macrophage migration inhibitory factor (MIF) gene is associated with adolescents' cortisol reactivity and anxiety. Psychoneuroendocrinology 2018; 95:170-178. [PMID: 29870971 DOI: 10.1016/j.psyneuen.2018.05.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 12/19/2022]
Abstract
Emerging evidence points to interactions between inflammatory markers and stress reactivity in predicting mental health risk, but underlying mechanisms are not well understood. Macrophage Migration Inhibitory Factor (MIF) is a pleiotropic cytokine involved in inflammatory signaling and Hypothalamus Pituitary Adrenal (HPA) axis stress-response, and has recently been identified as a candidate biomarker for depression and anxiety risk. We examined polymorphic variations of the MIF gene in association with baseline MIF levels, HPA axis reactivity, and self-reported anxiety responses to a social stressor in 74 adolescents, ages 10-14 years. Genotyping was performed for two polymorphisms, the -794 CATT5-8 tetranucleotide repeat and the -173*G/C single nucleotide polymorphism (SNP). Youth carrying the MIF-173*C and CATT7 alleles displayed attenuated cortisol reactivity when compared with non-carriers. Children with the CATT7-173*C haplotype displayed lower cortisol reactivity to the stressor compared to those without this haplotype. Additionally, the CATT5-173*C and CATT6-173*C haplotypes were associated with lower self-reported anxiety ratings across the stressor. Results extend prior work pointing to the influence of MIF signaling on neuroendocrine response to stress and suggest a potential pathophysiological pathway underlying risk for stress-related physical and mental health disorders. To our knowledge, these are the first data showing associations between the MIF gene, HPA axis reactivity, and anxiety symptoms during adolescence.
Collapse
Affiliation(s)
- Rebecca Lipschutz
- Department of Psychology, University of Houston, Houston, TX, United States
| | - Johanna Bick
- Department of Psychology, University of Houston, Houston, TX, United States.
| | - Victoria Nguyen
- Child Study Center, Yale School of Medicine, New Haven, CT, United States
| | - Maria Lee
- Child Study Center, Yale School of Medicine, New Haven, CT, United States
| | - Lin Leng
- Department of Internal Medicine, Rheumatology, Yale School of Medicine, New Haven, CT, United States
| | - Elena Grigorenko
- Department of Psychology, University of Houston, Houston, TX, United States; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Richard Bucala
- Department of Internal Medicine, Rheumatology, Yale School of Medicine, New Haven, CT, United States
| | - Linda C Mayes
- Child Study Center, Yale School of Medicine, New Haven, CT, United States
| | - Michael J Crowley
- Child Study Center, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
6
|
Zheng X, Li S, Zhang W, Zang Z, Hu J, Yang H. Current biomarkers of invasive sporadic pituitary adenomas. ANNALES D'ENDOCRINOLOGIE 2016; 77:658-667. [PMID: 27659267 DOI: 10.1016/j.ando.2016.02.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 01/29/2016] [Accepted: 02/21/2016] [Indexed: 12/22/2022]
Abstract
Though pituitary adenomas (PA) are considered benign, some of them exhibit invasive behaviors such as recurrence and low rate of total surgical resection. Reliable prognostic biomarkers for invasive PA are highly desired; however they remain to be identified. In this review, we summarize the current controversial findings of biomarkers for invasive sporadic PA, and we discuss the possible reasons for the controversies.
Collapse
Affiliation(s)
- Xin Zheng
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183, Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Song Li
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183, Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Weihua Zhang
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Zhenle Zang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183, Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Jintao Hu
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183, Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Hui Yang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183, Xinqiao Main Street, Shapingba District, Chongqing, China.
| |
Collapse
|
7
|
Koizumi M, Nahar A, Yamabe R, Kadokawa H. Positive correlations of age and parity with plasma concentration of macrophage migration inhibitory factor in Japanese black cows. J Reprod Dev 2016; 62:257-63. [PMID: 26853787 PMCID: PMC4919289 DOI: 10.1262/jrd.2015-144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Plasma Macrophage migration inhibitory factor (MIF) concentration correlates positively with age, and
negatively with self-rated health in women, and optimal MIF concentration may promote proper reproductive
function. This study was conducted to evaluate the hypotheses that plasma MIF concentration changes with
parturition or postpartum first ovulation, and that age in months and parity correlate with plasma MIF
concentration in Japanese black cows. Western blotting utilizing an anti-MIF mouse monoclonal antibody of
various tissues and plasma from females indicated that MIF expression was stronger in the anterior pituitary
than in other tissues. We developed a competitive EIA utilizing the same anti-MIF mouse monoclonal antibody
with sufficient sensitivity and reliable performance for measuring bovine plasma samples. We then measured MIF
concentrations in bovine plasma collected from 4 weeks before parturition to 4 weeks after postpartum first
ovulation. There was no significant difference in plasma MIF concentration pre- and post-parturition, or
before and after the postpartum first ovulation. Plasma MIF concentrations were positively correlated (P <
0.01) with parity (r = 0.703), age in months on the day of parturition (r = 0.647), and age in months on the
day of the postpartum first ovulation (r = 0.553) when we used almost all data, except for that from a
third-parity cow with an abnormally high plasma MIF concentration. We therefore concluded that plasma MIF
concentrations may increase with age in months and parity, but do not change either before and after
parturition or before and after postpartum first ovulation in Japanese black cows.
Collapse
Affiliation(s)
- Motoya Koizumi
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8515, Japan
| | | | | | | |
Collapse
|
8
|
Bick J, Nguyen V, Leng L, Piecychna M, Crowley MJ, Bucala R, Mayes LC, Grigorenko EL. Preliminary associations between childhood neglect, MIF, and cortisol: potential pathways to long-term disease risk. Dev Psychobiol 2015; 57:131-9. [PMID: 25380347 PMCID: PMC4337818 DOI: 10.1002/dev.21265] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 10/04/2014] [Indexed: 11/06/2022]
Abstract
The study examined Hypothalamus-Pituitary-Adrenal (HPA) axis and inflammatory signaling in 206 youth with histories of prenatal drug exposure and self-reported histories of maltreatment. Youth with histories of severe neglect showed elevated levels of cortisol, the end product of the HPA axis, in comparison to youth with lower or minimal levels of neglect. Histories of severe neglect also were associated with increased levels of Macrophage Migration Inhibitory Factor (MIF), a cytokine known to be intricately involved in HPA axis regulation. Salivary MIF levels also were positively associated with youth age and prenatal drug exposure. These MIF and cortisol alterations may signal pathophysiological disruptions in the neuro-endocrine and immune systems, which may lead to trajectories of increased disease risk among vulnerable youth. Our findings also provide preliminary support for the validity and reliability of a noninvasive salivary assessment of MIF.
Collapse
Affiliation(s)
- Johanna Bick
- Child Study Center, Yale School of Medicine, New Haven, CT
| | - Victoria Nguyen
- Child Study Center, Yale School of Medicine, New Haven, CT
- Yale College, New Haven, CT
| | - Lin Leng
- Department of Internal Medicine, Rheumatology, Yale School of Medicine, New Haven, CT
| | - Marta Piecychna
- Department of Internal Medicine, Rheumatology, Yale School of Medicine, New Haven, CT
| | | | - Richard Bucala
- Department of Internal Medicine, Rheumatology, Yale School of Medicine, New Haven, CT
| | - Linda C. Mayes
- Child Study Center, Yale School of Medicine, New Haven, CT
| | - Elena L. Grigorenko
- Child Study Center, Yale School of Medicine, New Haven, CT
- Moscow State University for Psychology, and Education, Moscow, Russia
| |
Collapse
|
9
|
Khalyfa A, Kheirandish-Gozal L, Capdevila OS, Bhattacharjee R, Gozal D. Macrophage migration inhibitory factor gene polymorphisms and plasma levels in children with obstructive sleep apnea. Pediatr Pulmonol 2012; 47:1001-11. [PMID: 22451332 PMCID: PMC3405200 DOI: 10.1002/ppul.22560] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2011] [Accepted: 11/28/2011] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Obstructive sleep apnea (OSA) is associated with increased risk for cardiovascular and metabolic dysfunction in both adults and children. In adults with OSA, serum levels of macrophage migration inhibitory factor (MIF) are elevated. Therefore, we assessed plasma MIF levels and MIF allelic variant frequencies in children with and without OSA (NOSA). METHODS A total of 614 consecutive children ages 5-8 years were recruited. Children were divided into those with OSA and NOSA based on the apnea-hypopnea index (AHI). In addition to lipid profile, hsCRP, and fasting insulin and glucose levels, plasma MIF levels were assayed using ELISA, and 28 single nucleotide polymorphisms (SNPs) covering the region were genotyped. Linkage disequilibrium and haplotype blocks were analyzed using Haploview version 4.2 software. RESULTS Morning plasma MIF levels were increased in children with OSA. Of the 28 SNPs tested, the frequency of rs10433310 minor allele was significantly decreased in OSA. This SNP was also associated with reduced fasting insulin and hsCRP levels in OSA. The minor allele frequency of all other 27 SNPs was similar in OSA and NOSA groups. CONCLUSIONS Childhood OSA is associated with higher plasma MIF, hsCRP, and fasting insulin levels that promote cardiometabolic risk, and the MIF gene SNP rs10433310 may account for some of the variance in such risk.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Department of Pediatrics, Comer Children's Hospital, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | |
Collapse
|
10
|
Musil R, Schwarz MJ, Riedel M, Dehning S, Cerovecki A, Spellmann I, Arolt V, Müller N. Elevated macrophage migration inhibitory factor and decreased transforming growth factor-beta levels in major depression--no influence of celecoxib treatment. J Affect Disord 2011; 134:217-25. [PMID: 21684012 DOI: 10.1016/j.jad.2011.05.047] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 05/26/2011] [Accepted: 05/26/2011] [Indexed: 12/22/2022]
Abstract
OBJECTIVES The involvement of an immune process in the pathophysiology of major depression disorder (MDD) was substantiated by studies demonstrating elevated levels of proinflammatory cytokines and prostaglandin E(2) (PGE(2)). Cyclooxygenase-2 (COX-2) inhibitors lead to a reduced production of PGE(2) and have been shown to improve depressive symptoms. We investigated the three immune parameters macrophage migration inhibitory factor (MIF), transforming growth factor-β (TGF-β) and soluble CD14 (sCD14) in a randomized, placebo-controlled trial of the COX-2 inhibitor celecoxib as add-on therapy in patients with MDD treated with reboxetine. METHODS Thirty-two patients with depression and 20 healthy controls participated in the study. The patients were treated with reboxetine and celecoxib or placebo. Immune parameters were measured from serum at baseline, after three and five weeks using ELISA. RESULTS Celecoxib as add-on strategy resulted in a significant reduction of Hamilton Depression Scale scores compared to placebo. Depressed patients showed significantly elevated MIF (p < 0.001) and reduced TGF-β (p = 0.006) concentrations at baseline. There was no difference in sCD14-concentrations. There was no difference between the placebo and the celecoxib group and no change over time. LIMITATIONS Limitations of the study are the relatively small sample size and lack of functional assessment of HPA axis in parallel. CONCLUSIONS MIF is a promising new candidate in the neuro-immune interplay that may link depressive symptoms, altered immune state and HPA-axis dysregulation. Reduced levels of TGF-β replicate previous findings and support the importance of this regulatory cytokine in major depressive disorder.
Collapse
Affiliation(s)
- R Musil
- Psychiatric Clinic of University Munich, Nussbaumstrasse 7, 80336 Munich, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Elevated macrophage migration inhibitory factor (MIF) is associated with depressive symptoms, blunted cortisol reactivity to acute stress, and lowered morning cortisol. Brain Behav Immun 2010; 24:1202-8. [PMID: 20382217 DOI: 10.1016/j.bbi.2010.03.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2009] [Revised: 03/31/2010] [Accepted: 03/31/2010] [Indexed: 11/24/2022] Open
Abstract
Macrophage Migration Inhibitory Factor (MIF) is a proinflammatory cytokine produced by leukocytes and the secretory cells of the HPA axis. Remarkably, glucocorticoids (GC) induce leukocyte MIF secretion, while MIF renders leukocytes insensitive to the anti-inflammatory effects of glucocorticoids. In light of reported associations between dysphoric states, increased inflammatory activity, and reduced GC sensitivity, the current study investigated the association between MIF, loneliness and depressive symptoms. The study further investigated the relation between plasma MIF and markers of HPA function, i.e., diurnal cortisol and the cortisol response to acute stress. Healthy university undergraduates (N=126; 64 women) were invited to participate if their scores on the Beck Depression Inventory or UCLA loneliness scale were in the upper or lower quintile of their peer group. Plasma MIF and salivary cortisol were measured in response to a public speaking task. Ambulatory diurnal cortisol was assessed for 5 consecutive days. MIF levels were 40% higher in the high-depressive symptoms group compared to the low depressive symptoms group. Elevated MIF was also associated with a smaller cortisol response to acute stress and lower diurnal morning cortisol values. The observed association between HPA function and MIF remained robust after adjustment for depressive symptoms, and demographic, anthropomorphic, and behavioural factors. High levels of depressive symptoms were likewise associated with lower morning cortisol, but this association became non-significant after adjustment for MIF. MIF may be an important neuro-immune mediator linking depressive symptoms with inflammation and HPA dysregulation.
Collapse
|
12
|
Rees DA, Giles P, Lewis MD, Ham J. Adenosine regulates thrombomodulin and endothelial protein C receptor expression in folliculostellate cells of the pituitary gland. Purinergic Signal 2010; 6:19-29. [PMID: 19859827 PMCID: PMC2837818 DOI: 10.1007/s11302-009-9172-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Accepted: 10/08/2009] [Indexed: 12/21/2022] Open
Abstract
Adenosine stimulates the release of interleukin 6 (IL-6) and vascular endothelial growth factor from folliculostellate cells of the anterior pituitary gland indicating that such cells are also involved in the communication between the immune and endocrine systems during stress and inflammation. In order to understand the precise actions of adenosine on folliculostellate cells, DNA microarray analysis was used to determine global changes in gene expression. Hierarchical clusters revealed, of the genes that had altered expression, the majority were suppressed and many, such as B cell translocation gene 2 and cyclin-dependent kinase inhibitor 2b were related to cell cycle arrest or inhibition of proliferation. Several of the up-regulated genes were associated with cytokine signalling or membrane receptor activity. The most notable of these being IL-6, sulfiredoxin 1, endothelial protein C receptor (EPCR) and thrombomodulin (THBD) which can all play a role in controlling inflammation. The EPCR and THBD pathway is well known in anti-coagulation but also has anti-inflammatory and anti-apoptotic properties. Up-regulation of EPCR and THBD in folliculostellate cells was confirmed by qRT-PCR and western blotting analysis and their expression were also demonstrated in many of the hormone-secreting cells of the anterior pituitary gland. Our findings suggest that adenosine can stimulate expression of stress and inflammation related genes from folliculostellate cells of the anterior pituitary gland. These genes include EPCR and THBD, neither of which has been previously identified in the pituitary gland.
Collapse
Affiliation(s)
- D. Aled Rees
- Centre for Endocrine and Diabetes Sciences, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN UK
| | - Peter Giles
- Department of Pathology, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN UK
| | - Mark D. Lewis
- Centre for Endocrine and Diabetes Sciences, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN UK
| | - Jack Ham
- Centre for Endocrine and Diabetes Sciences, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN UK
| |
Collapse
|
13
|
Wang F, Shen X, Guo X, Peng Y, Liu Y, Xu S, Yang J. Spinal macrophage migration inhibitory factor contributes to the pathogenesis of inflammatory hyperalgesia in rats. Pain 2009; 148:275-283. [PMID: 20005040 DOI: 10.1016/j.pain.2009.11.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Revised: 10/28/2009] [Accepted: 11/10/2009] [Indexed: 12/12/2022]
Abstract
Pro-inflammatory cytokine production after nociceptive stimuli is pivotal for hyperalgesia. As macrophage migration inhibitory factor (MIF), a pleiotropic cytokine produced mainly by nonneuronal tissue, has been involved in the regulation of neuronal functions, herein we examined the role for MIF in formalin-induced inflammatory pain model. MIF critically contributed to nociceptive behaviors following formalin injection. Specifically, spinal administration of a MIF inhibitor (ISO-1) prevented and reversed flinching responses in rats. Further examination showed that levels of both MIF and the MIF receptor CD74 were substantially increased within the ipsilateral spinal cord dorsal horn after formalin administration. Mechanistic studies revealed that MIF upregulated the expression of the spinal NMDA receptor subunit NR2B via the MAPK signaling pathway. Moreover, microglial cells were found to be the major source of spinal MIF after formalin administration by fluorescence colocalization. These data highlight spinal MIF plays a critical role in the pathogenesis of formalin-induced inflammatory pain and suggest MIF may be a potential target for therapy of such pathological condition.
Collapse
Affiliation(s)
- FuZhou Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, No. 22, Hankou Road, Nanjing 210093, Jiangsu, China Department of Anesthesiology, The Affiliated Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing 210004, Jiangsu, China The Institute of Pediatrics, The Affiliated Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing 210004, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|
14
|
Sánchez-Lemus E, Benicky J, Pavel J, Saavedra JM. In vivo Angiotensin II AT1 receptor blockade selectively inhibits LPS-induced innate immune response and ACTH release in rat pituitary gland. Brain Behav Immun 2009; 23:945-57. [PMID: 19427376 PMCID: PMC2749886 DOI: 10.1016/j.bbi.2009.04.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Revised: 04/20/2009] [Accepted: 04/30/2009] [Indexed: 12/23/2022] Open
Abstract
Systemic lipopolysaccharide (LPS) administration induces an innate immune response and stimulates the hypothalamic-pituitary-adrenal axis. We studied Angiotensin II AT(1) receptor participation in the LPS effects with focus on the pituitary gland. LPS (50 microg/kg, i.p.) enhanced, 3h after administration, gene expression of pituitary CD14 and that of Angiotensin II AT(1A) receptors in pituitary and hypothalamic paraventricular nucleus (PVN); stimulated ACTH and corticosterone release; decreased pituitary CRF(1) receptor mRNA and increased all plasma and pituitary pro-inflammatory factors studied. The AT(1) receptor blocker (ARB) candesartan (1mg/kg/day, s.c. daily for 3 days before LPS) blocked pituitary and PVN AT(1) receptors, inhibited LPS-induced ACTH but not corticosterone secretion and decreased LPS-induced release of TNF-alpha, IL-1beta and IL-6 to the circulation. The ARB reduced LPS-induced pituitary gene expression of IL-6, LIF, iNOS, COX-2 and IkappaB-alpha; and prevented LPS-induced increase of nNOS/eNOS activity. The ARB did not affect LPS-induced TNF-alpha and IL-1beta gene expression, IL-6 or IL-1beta protein content or LPS-induced decrease of CRF(1) receptors. When administered alone, the ARB increased basal plasma corticosterone levels and basal PGE(2) mRNA in pituitary. Our results demonstrate that the pituitary gland is a target for systemically administered LPS. AT(1) receptor activity is necessary for the complete pituitary response to LPS and is limited to specific pro-inflammatory pathways. There is a complementary and complex influence of the PVN and circulating cytokines on the initial pituitary response to LPS. Our findings support the proposal that ARBs may be considered for the treatment of inflammatory conditions.
Collapse
Affiliation(s)
- Enrique Sánchez-Lemus
- Section on Pharmacology, Division of Intramural Research Programs, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
15
|
Leng L, Wang W, Roger T, Merk M, Wuttke M, Calandra T, Bucala R. Glucocorticoid-induced MIF expression by human CEM T cells. Cytokine 2009; 48:177-85. [PMID: 19646897 DOI: 10.1016/j.cyto.2009.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 05/07/2009] [Accepted: 07/06/2009] [Indexed: 01/01/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is an upstream activator of the immune response that counter-regulates the immunosuppressive effects of glucocorticoids. While MIF is released by cells in response to diverse microbial and invasive stimuli, evidence that glucocorticoids in low concentrations also induce MIF secretion suggests an additional regulatory relationship between these mediators. We investigated the expression of MIF from the human CEM T cell line, which exists in two well-characterized, glucocorticoid-sensitive (CEM-C7) and glucocorticoid-resistant (CEM-C1) variant clones. Dexamethasone in low concentrations induced MIF secretion from CEM-C7 but not CEM-C1 T cells by a bell-shaped dose response that was similar to that reported previously for the release of MIF by monocytes/macrophages. Glucocorticoid stimulation of CEM-C7 T cells was accompanied by an MIF transcriptional response, which by promoter analysis was found to involve the GRE and ATF/CRE transcription factor binding sites. These data support a glucocorticoid-mediated MIF secretion response by T cells that may contribute to the regulation of the adaptive immune response.
Collapse
Affiliation(s)
- Lin Leng
- Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
Folliculo-stellate cells (FS-cells) are star-shaped and follicle-forming cells in the anterior pituitary gland that were first identified by electron microscopy as non-endocrine agranular cells. Light microscopy has revealed many of their cytophysiological features and the FS-cell is known to be positive for S-100 protein, a marker for FS-cells. So far, functions ascribed to FS-cells include the formation of an extensive and complex tridimentional network, scavenger activity by engulfing degenerated cells, paracrine regulation of endocrine cells by producing various growth factors and cytokines, such as interleukin-6, leukemia inhibitory factor, basic fibroblastic growth factor, vascular endothelial cell growth factor and follistatin, and large-scale inter-cellular communication by means of their long cytoplasmic processes and gap junctions. Moreover, their multi-potential characteristics and other cytological features support the possibility of them becoming organ-specific stem cells. This concept is yet to be resolved, however. In this review, we focus on these features of FS-cells along with some futuristic approaches.
Collapse
Affiliation(s)
- S Devnath
- Department of Regulation Biology, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | | |
Collapse
|
17
|
Bernstein IL, Li JT, Bernstein DI, Hamilton R, Spector SL, Tan R, Sicherer S, Golden DBK, Khan DA, Nicklas RA, Portnoy JM, Blessing-Moore J, Cox L, Lang DM, Oppenheimer J, Randolph CC, Schuller DE, Tilles SA, Wallace DV, Levetin E, Weber R. Allergy diagnostic testing: an updated practice parameter. Ann Allergy Asthma Immunol 2008; 100:S1-148. [PMID: 18431959 DOI: 10.1016/s1081-1206(10)60305-5] [Citation(s) in RCA: 291] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
18
|
Effects of glucocorticoid agonist and antagonist on the pathogenesis of L-arginine-induced acute pancreatitis in rat. Pancreas 2008; 36:369-76. [PMID: 18437083 DOI: 10.1097/mpa.0b013e31815bd26a] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVES To investigate the consequences of treatment with an exogenous glucocorticoid agonist (methylprednisolone) and antagonist (RU-38486) on the local and systemic responses in L-arginine-induced acute pancreatitis in rats. METHODS The methylprednisolone and RU-38486 were administered just before pancreatitis induction. Plasma amylase activity, interleukin 6 activity, pancreatic weight/body weight ratio, plasma macrophage migration inhibitory factor (MIF) concentration, and pancreatic nuclear transcription factor (NF) kappaB activity were determined. The extents of pancreas, liver, and lung injuries were assessed by histology. RESULTS Acute pancreatitis resulted in NF-kappaB activation and proinflammatory cytokine release in rats. In the glucocorticoid agonist group, plasma amylase and interleukin 6 levels were significantly decreased as compared with those of RU-38486 and nontreated groups. Antagonist treatment led to significantly higher MIF production at 8 and 12 hours after L-arginine injection as compared with the agonist-treated and nontreated groups. Glucocorticoid agonist treatment significantly decreased the level of NF-kappaB 24 hours after pancreatitis induction. Histological investigations showed protective effect of agonist treatment on acute pancreatitis-induced tissue damage in the pancreas and lung. CONCLUSIONS These results corroborated the importance of MIF in acute pancreatitis. The glucocorticoid-dependent mechanisms seem to play a crucial role in the control of the inflammatory response and tissue damage in L-arginine-induced experimental acute pancreatitis.
Collapse
|
19
|
Cunning factor: macrophage migration inhibitory factor as a redox‐regulated target. Immunol Cell Biol 2007; 86:232-8. [DOI: 10.1038/sj.icb.7100133] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
20
|
John CD, Theogaraj E, Christian HC, Morris JF, Smith SF, Buckingham JC. Time-specific effects of perinatal glucocorticoid treatment on anterior pituitary morphology, annexin 1 expression and adrenocorticotrophic hormone secretion in the adult female rat. J Neuroendocrinol 2006; 18:949-59. [PMID: 17076770 DOI: 10.1111/j.1365-2826.2006.01493.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Perinatal glucocorticoid (GC) treatment is increasingly associated with long-term disturbances in hypothalamo-pituitary-adrenocortical function. In the male rat, such treatment induces profound molecular, morphological and functional changes in the anterior pituitary gland at adulthood. To determine whether these effects are sex-specific, we have examined the effects of perinatal dexamethasone treatment on the female pituitary gland, focusing on (i) the integrity of the annexin 1 (ANXA1) dependent regulatory effects of GCs on adrenocorticotrophic hormone (ACTH) release and (ii) corticotroph and folliculo-stellate (FS) cell morphology. Dexamethasone was given to pregnant (gestational days 16-19) or lactating (days 1-7 post partum) rats via the drinking water (1 microg/ml); controls received normal drinking water. Pituitary tissue from the female offspring was examined ex vivo at adulthood (60-90 days). Both treatment regimes reduced the intracellular and cell surface ANXA1 expression, as determined by western blot analysis and quantitative immunogold electron microscopic histochemistry. In addition, they compromised the ability of dexamethasone to suppress the evoked release of ACTH from the excised tissue in vitro, a process which requires the translocation of ANXA1 from the cytoplasm to the cell surface of FS cells. Although neither treatment regime affected the number of FS cells or corticotrophs, both altered the subcellular morphology of these cells. Thus, prenatal dexamethasone treatment increased while neonatal treatment decreased FS cell size and cytoplasmic area. By contrast, corticotroph size was unaffected by either treatment, as also was the size of the secretory granules. Corticotroph granule density and margination were, however, increased markedly by the prenatal treatment, while the neonatal treatment had no effect on granule density but decreased granule margination. Thus, perinatal dexamethasone treatment exerts long-term effects on the female pituitary gland, altering gene expression, cell morphology and the ANXA1-dependent GC regulation of ACTH secretion. The changes are similar but not identical to those reported in the male.
Collapse
Affiliation(s)
- C D John
- Department of Cellular and Molecular Neuroscience, Division of Neuroscience and Mental Health, Imperial College London, Hammersmith Campus, London, UK
| | | | | | | | | | | |
Collapse
|
21
|
Morris JF, Omer S, Davies E, Wang E, John C, Afzal T, Wain S, Buckingham JC, Flower RJ, Christian HC. Lack of annexin 1 results in an increase in corticotroph number in male but not female mice. J Neuroendocrinol 2006; 18:835-46. [PMID: 17026533 PMCID: PMC1855440 DOI: 10.1111/j.1365-2826.2006.01481.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Annexin 1 (ANXA1) is a member of the annexin family of phospholipid- and calcium-binding proteins with a well demonstrated role in early delayed (30 min to 3 h) inhibitory feedback of glucocorticoids in the pituitary. We have examined corticotrophs in wild-type and ANXA1 knockout mice to determine the effects of lack of ANXA1 in male and female animals. Anterior pituitary tissue from ANXA1 wild-type, heterozygote and null mice was fixed and examined (i) by confocal immunocytochemistry to determine the number of corticotrophs and (ii) by electron microscopy to examine the size, secretory granule population and secretory machinery of corticotrophs. No differences in these parameters were detected in female mice. In male ANXA1 null mice, there were approximately four-fold more corticotrophs than in wild-type animals. However, the corticotrophs in ANXA1 null mice were smaller and had reduced numbers of secretory granules (the reduction in granules paralleled the reduction in cell size). No differences in the numerical density of folliculo-stellate, gonadotroph, lactotroph or somatotroph cells were detected in male ANXA1 null mice. Plasma corticosterone, adrenocorticotrophic hormone (ACTH) and pituitary pro-opiomelanocortin mRNA were unchanged but pituitary ACTH content was increased in male ANXA1 null mice. Interleukin (IL)-6 pituitary content was significantly elevated in male and reduced in female ANXA1 null mice compared to wild-type. In conclusion, these data indicate that ANXA1 deficiency is associated with gender-specific changes in corticotroph number and structure, via direct actions of ANXA1 and/or indirect changes in factors such as IL-6.
Collapse
Affiliation(s)
- J F Morris
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Kudrin A, Scott M, Martin S, Chung CW, Donn R, McMaster A, Ellison S, Ray D, Ray K, Binks M. Human macrophage migration inhibitory factor: a proven immunomodulatory cytokine? J Biol Chem 2006; 281:29641-51. [PMID: 16893895 DOI: 10.1074/jbc.m601103200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pro-inflammatory mediator with the ability to induce various immunomodulatory responses and override glucocorticoid-driven immunosuppression. Some of these functions have been linked to the unusual enzymatic properties of the protein, namely tautomerase and oxidoreductase activities. However, there are conflicting reports regarding the functional role of these enzymatic properties in normal physiological homeostasis and disease progression. Therefore, we have produced a highly pure, virtually endotoxin-free recombinant MIF preparation and fully characterized this using a variety of biochemical and biophysical approaches. The recombinant protein, with demonstrable enzymatic activity, was then used to systematically examine the biological activity of MIF. Surprisingly, treatment with MIF alone failed to induce cytokine expression, with the exception of IL-8. However, co-treatment of lipopolysaccharide (LPS) in conjunction with MIF produced synergistic secretion of tumor necrosis factor-alpha, interleukin (IL)-1, and IL-8 compared with LPS alone. The potentiating effect of MIF was seen at physiologically relevant concentrations. These data suggest that MIF has no conventional cytokine activity but, rather, acts to modulate and amplify the response to LPS.
Collapse
Affiliation(s)
- Alex Kudrin
- Department of Disease Biology, Rheumatology, and Inflammation and Discovery Research, GlaxoSmithKline, Stevenage SG1 2NY, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Bruhn A, Verdant C, Vercruysse V, Su F, Vray B, Vincent JL. EFFECTS OF DEXAMETHASONE ON MACROPHAGE MIGRATION INHIBITORY FACTOR PRODUCTION IN SEPSIS. Shock 2006; 26:169-73. [PMID: 16878025 DOI: 10.1097/01.shk.0000225416.27742.cb] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine that plays a major role in the pathogenesis of sepsis. Some studies have indicated that glucocorticoids increase MIF production in physiological conditions. The goal of this study was to determine whether glucocorticoid treatment also upregulates MIF production in sepsis. Male NMRI mice (6-10 weeks old) underwent laparotomy, proximal ligation of the cecum, and double perforation with a 19-gauge needle (cecal ligation and puncture). Mice were randomly treated with saline (control) or dexamethasone at doses of 0.1, 1, or 10 mg/kg ip. At 6 or 18 h postoperatively, 10 mice per group were euthanized; and blood, peritoneal fluid, liver, lung, kidney, and heart tissue samples were retrieved. MIF, IL-6, TNF-alpha, and IL-10 were measured by enzyme-linked immunosorbent assay. Sepsis induced by cecal ligation and puncture produced a marked increase in MIF and cytokine levels in plasma and peritoneal fluid. Treatment with dexamethasone 10 mg/kg decreased MIF levels in plasma after 18 h, but there was no effect of dexamethasone on MIF production locally in the peritoneal cavity or in the liver, lungs, heart, or kidneys. We conclude that glucocorticoid treatment does not upregulate MIF production in sepsis.
Collapse
|
24
|
Abstract
The cytokine macrophage migration inhibitory factor (MIF) participates in fundamental events in innate and adaptive immunity. The profile of activities of MIF in vivo and in vitro is strongly suggestive of a role for MIF in the pathogenesis of many inflammatory diseases, including rheumatoid arthritis (RA), and hence antagonism of MIF is suggested as a potential therapeutic strategy in inflammatory disease. The best developed case for therapeutic antagonism of MIF is in RA. In RA, MIF is abundantly expressed in serum and synovial tissue. MIF induces synovial expression of key pro-inflammatory genes, regulates the function of endothelial cells and leucocytes, and is implicated in the control of synoviocyte proliferation and apoptosis via direct effects on the expression of the tumour suppressor protein p53. In animal models of RA, anti-MIF antibodies or genetic MIF deficiency are associated with significant inhibition of disease. A similar case has been made, for example using MIF-deficient mice, in models of atheroma, colitis, multiple sclerosis and other inflammatory diseases. The relationship with p53 also means MIF may be important in the link between inflammatory disease and cancer, such as is seen in RA or colitis. MIF also has a unique relationship with glucocorticoids, in that despite antagonizing their effects, the expression of MIF is in fact induced by glucocorticoids. Thus, MIF functions as a physiological counter-regulator of the anti-inflammatory effects of glucocorticoids. This may be entrained by selective activation of mitogen-activated protein kinases rather than nuclear factor kappa B. Therapeutic MIF antagonism may therefore provide a specific means of 'steroid sparing'. Exploitation of antibody, soluble receptor or small molecule technologies may soon lead to the ability to test in the clinic the importance of MIF in human inflammatory diseases.
Collapse
Affiliation(s)
- E F Morand
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Melbourne, Victoria, Australia.
| |
Collapse
|
25
|
van der Hoek J, Waaijers M, van Koetsveld PM, Sprij-Mooij D, Feelders RA, Schmid HA, Schoeffter P, Hoyer D, Cervia D, Taylor JE, Culler MD, Lamberts SWJ, Hofland LJ. Distinct functional properties of native somatostatin receptor subtype 5 compared with subtype 2 in the regulation of ACTH release by corticotroph tumor cells. Am J Physiol Endocrinol Metab 2005; 289:E278-87. [PMID: 15769796 DOI: 10.1152/ajpendo.00004.2005] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In a series of human corticotroph adenomas, we recently found predominant mRNA expression of somatostatin (SS) receptor subtype 5 (sst5). After 72 h, the multiligand SS analog SOM230, which has a very high sst5 binding affinity, but not Octreotide (OCT), significantly inhibited basal ACTH release. To further explore the role of sst5 in the regulation of ACTH release, we conducted additional studies with mouse AtT-20 cells. SOM230 showed a 7-fold higher ligand binding affinity and a 19-fold higher potency in stimulating guanosine 5'-O-(3-thiotriphosphate) binding in AtT-20 cell membranes compared with OCT. SOM230 potently suppressed CRH-induced ACTH release, which was not affected by 48-h dexamethasone (DEX) pretreatment. However, DEX attenuated the inhibitory effects of OCT on ACTH release, whereas it increased the inhibitory potency of BIM-23268, an sst5-specific analog, on ACTH release. Quantitative PCR analysis showed that DEX lowered sst(2A+2B) mRNA expression significantly after 24 and 48 h, whereas sst5 mRNA levels were not significantly affected by DEX treatment. Moreover, Scatchard analyses showed that DEX suppressed maximum binding capacity (B(max)) by 72% when 125I-Tyr3-labeled OCT was used as radioligand, whereas B(max) declined only by 17% when AtT-20 cells were treated with [125I-Tyr11]SS-14. These data suggest that the sst5 protein, compared with sst2, is more resistant to glucocorticoids. Finally, after SS analog preincubation, compared with OCT both SOM230 and BIM-23268 showed a significantly higher inhibitory effect on CRH-induced ACTH release. In conclusion, our data support the concept that the sst5 receptor might be a target for new therapeutic agents to treat Cushing's disease.
Collapse
Affiliation(s)
- Joost van der Hoek
- Dept. of Internal Medicine, section Endocrinology, Erasmus MC, 3015 GE Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Historically, the study of folliculo-stellate (FS) cells of the anterior pituitary dates back to the onset of electron microscopical observation of the pituitary gland. The morphological and electrophysiological characteristics, topographical distribution and contribution to intercellular junctions of these FS cells have been instrumental to the understanding of their putative function. Moreover, many studies have documented the role of FS cells as a source of newly discovered peptides, growth factors and cytokines. Quantitative immunohistochemical observation of FS cells in situ and functional in vitro studies, using either cultured FS cells or cells from an immortalized FS cell line, forwarded the notion of immunophenotypical and functional heterogeneity of the FS cell group. Double immunolabeling with a classical FS cell marker (S-100 protein) and with major histocompatibility complex class II markers characteristic for dendritic cells (DC) have shown a considerable overlap of FS cells with DC. The latter cells are immunocompetent cells belonging to the mononuclear phagocyte system. In this review, the FS cell heterogeneity is discussed with respect to the question of their embryological origin and developmental fate and with respect to the physiological relevance of functionally heterogeneous subpopulations. Recent findings of a myeloid origin of part of the interstitial cells of the anterior pituitary are confronted by other developmental paradigms of pituitary cell differentiation. The possibility that FS cells represent an adult stem cell population of the pituitary is critically examined. Also the physiological role of FS cells in the interferon-gamma- and nitric oxide-mediated effects on pituitary hormone secretion is discussed. New approaches for the study of this enigmatic cell group using immortalized cell lines and new markers for an hitherto unrecognized pituitary cell population, the so-called 'side population', are evaluated.
Collapse
Affiliation(s)
- Wilfried Allaerts
- Biological Publishing, PO Box 104, NL-7440 AC Nijverdal, The Netherlands.
| | | |
Collapse
|
27
|
Herkenham M. Folliculo-stellate (FS) cells of the anterior pituitary mediate interactions between the endocrine and immune systems. Endocrinology 2005; 146:33-4. [PMID: 15601904 DOI: 10.1210/en.2004-1269] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|