1
|
Bhardwaj I, Singh S, Ansari AH, Rai SP, Singh D. Effect of stress on neuronal cell: Morphological to molecular approach. PROGRESS IN BRAIN RESEARCH 2025; 291:469-502. [PMID: 40222791 DOI: 10.1016/bs.pbr.2025.01.010] [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: 04/15/2025]
Abstract
Stress can be characterized as any perceived or actual threat that necessitates compensatory actions to maintain homeostasis. It can alter an organism's behavior over time by permanently altering the composition and functionality of brain circuitry. The amygdala and prefrontal cortex are two interrelated brain regions that have been the focus of initial research on stress and brain structural and functional plasticity, with the hippocampus serving as the entry point for most of this knowledge. Prolonged stress causes significant morphological alterations in important brain regions such as the hippocampus, amygdala, and prefrontal cortex. Memory, learning, and emotional regulation are among the cognitive functions that are adversely affected by these changes, including neuronal shrinkage, dendritic retraction, and synaptic malfunction. Stress perturbs the equilibrium of neurotransmitters, neuronal plasticity, and mitochondrial function at the molecular level. On the other hand, chronic stress negatively impacts physiology and can result in neuropsychiatric diseases. Recent molecular research has linked various epigenetic processes, such as DNA methylation, histone modifications, and noncoding RNAs, to the dysregulation of genes in the impacted brain circuits responsible for the pathophysiology of chronic stress. Numerous disorders, including neurodegenerative diseases (NDDs) including Alzheimer's, amyotrophic lateral sclerosis, Friedreich's ataxia, Huntington's disease, multiple sclerosis, and Parkinson's disease, have been linked to oxidative stress as a possible cause.
Collapse
Affiliation(s)
- Ishita Bhardwaj
- Department of Zoology, S.S. Khanna Girls' Degree College, Prayagraj (A Constituent College of University of Allahabad, Prayagraj), Uttar Pradesh, India
| | - Sippy Singh
- Department of Zoology, S.S. Khanna Girls' Degree College, Prayagraj (A Constituent College of University of Allahabad, Prayagraj), Uttar Pradesh, India
| | - Atifa Haseeb Ansari
- Department of Zoology, S.S. Khanna Girls' Degree College, Prayagraj (A Constituent College of University of Allahabad, Prayagraj), Uttar Pradesh, India
| | - Swayam Prabha Rai
- Department of Zoology, S.S. Khanna Girls' Degree College, Prayagraj (A Constituent College of University of Allahabad, Prayagraj), Uttar Pradesh, India
| | - Durgesh Singh
- Department of Zoology, S.S. Khanna Girls' Degree College, Prayagraj (A Constituent College of University of Allahabad, Prayagraj), Uttar Pradesh, India.
| |
Collapse
|
2
|
Schallmayer E, Isigkeit L, Elson L, Müller S, Knapp S, Marschner JA, Merk D. Chemogenomics for steroid hormone receptors (NR3). Commun Chem 2025; 8:29. [PMID: 39900826 PMCID: PMC11790914 DOI: 10.1038/s42004-025-01427-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025] Open
Abstract
The nine human NR3 nuclear receptors translate steroid hormone signals in transcriptomic responses and operate multiple highly important processes ranging from development over reproductive tissue function to inflammatory and metabolic homeostasis. Although several NR3 ligands such as glucocorticoids are invaluable drugs, this family is only partially explored, for example, in autoimmune diseases and neurodegeneration, but may hold therapeutic potential in new areas. Here we report a chemogenomics (CG) library to reveal elusive effects of NR3 receptor modulation in phenotypic settings. 34 highly annotated and chemically diverse ligands covering all NR3 receptors were selected considering complementary modes of action and activity, selectivity and lack of toxicity. Endoplasmic reticulum stress resolving effects of N3 CG subsets in proof-of-concept application validate suitability of the set to connect phenotypic outcomes with targets and to explore NR3 receptors from a translational perspective.
Collapse
Affiliation(s)
- Espen Schallmayer
- Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Laura Isigkeit
- Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Lewis Elson
- Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Susanne Müller
- Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Julian A Marschner
- Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Frankfurt, Germany.
- Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
3
|
Chen Z, Liu Y, Lyu M, Chan CH, Sun M, Yang X, Qiao S, Chen Z, Yu S, Ren M, Lu A, Zhang G, Li F, Yu Y. Classifications of triple-negative breast cancer: insights and current therapeutic approaches. Cell Biosci 2025; 15:13. [PMID: 39893480 PMCID: PMC11787746 DOI: 10.1186/s13578-025-01359-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/28/2025] [Indexed: 02/04/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and challenging type of cancer, characterized by the absence of specific receptors targeted by current therapies, which limits effective targeted treatment options. TNBC has a high risk of recurrence and distant metastasis, resulting in lower survival rates. Additionally, TNBC exhibits significant heterogeneity at histopathological, proteomic, transcriptomic, and genomic levels, further complicating the development of effective treatments. While some TNBC subtypes may initially respond to chemotherapy, resistance frequently develops, increasing the risk of aggressive recurrence. Therefore, precisely classifying and characterizing the distinct features of TNBC subtypes is crucial for identifying the most suitable molecular-based therapies for individual patients. In this review, we provide a comprehensive overview of these subtypes, highlighting their unique profiles as defined by various classification systems. We also address the limitations of conventional therapeutic approaches and explore innovative biological strategies, all aimed at advancing the development of targeted and effective therapeutic strategies for TNBC.
Collapse
Affiliation(s)
- Ziqi Chen
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, SAR, China
| | - Yumeng Liu
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, SAR, China
| | - Minchuan Lyu
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, SAR, China
| | - Chi Ho Chan
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, SAR, China
- Institute of Integrated Bioinformedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Meiheng Sun
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, SAR, China
| | - Xin Yang
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, SAR, China
- Institute of Integrated Bioinformedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Shuangying Qiao
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, SAR, China
| | - Zheng Chen
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, SAR, China
| | - Sifan Yu
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, SAR, China
- Institute of Integrated Bioinformedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Meishen Ren
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, People's Republic of China
| | - Aiping Lu
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, SAR, China
- Institute of Integrated Bioinformedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Ge Zhang
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, SAR, China
- Institute of Integrated Bioinformedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Fangfei Li
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, SAR, China
- Institute of Integrated Bioinformedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Yuanyuan Yu
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China.
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong, SAR, China.
- Institute of Integrated Bioinformedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China.
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China.
| |
Collapse
|
4
|
Amanollahi R, Holman SL, Bertossa MR, Meakin AS, Thornburg KL, McMillen IC, Wiese MD, Lock MC, Morrison JL. Ontogeny of Fetal Cardiometabolic Pathways: The Potential Role of Cortisol and Thyroid Hormones in Driving the Transition from Preterm to Near-Term Heart Development in Sheep. J Cardiovasc Dev Dis 2025; 12:36. [PMID: 39997470 PMCID: PMC11856455 DOI: 10.3390/jcdd12020036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/30/2024] [Accepted: 01/13/2025] [Indexed: 02/26/2025] Open
Abstract
Understanding hormonal and molecular changes during the transition from preterm to near-term gestation is essential for investigating how pregnancy complications impact fetal heart development and contribute to long-term cardiovascular risks for offspring. This study examines these cardiac changes in fetal sheep, focusing on the changes between 116 days (preterm) and 140 days (near term) of gestation (dG, term = 150) using Western blotting, LC-MS/MS, and histological techniques. We observed a strong correlation between cortisol and T3 (Triiodothyronine) in heart tissue in near-term fetuses, highlighting the role of glucocorticoid signalling in fetal heart maturation. Protein expression patterns in the heart revealed a decrease in multiple glucocorticoid receptor isoforms (GRα-A, GR-P, GR-A, GRα-D2, and GRα-D3), alongside a decrease in IGF-1R (a marker of cardiac proliferative capacity) and p-FOXO1(Thr24) but an increase in PCNA (a marker of DNA replication), indicating a shift towards cardiomyocyte maturation from preterm to near term. The increased expression of proteins regulating mitochondrial biogenesis and OXPHOS complex 4 reflects the known transition from glycolysis to oxidative phosphorylation, essential for meeting the energy demands of the postnatal heart. We also found altered glucose transporter expression, with increased pIRS-1(ser789) and GLUT-4 but decreased GLUT-1 expression, suggesting improved insulin responsiveness as the heart approaches term. Notably, the reduced protein abundance of SIRT-1 and SERCA2, along with increased phosphorylation of cardiac Troponin I(Ser23/24), indicates adaptations for more energy-efficient contraction in the near-term heart. In conclusion, these findings show the complex interplay of hormonal, metabolic, and growth changes that regulate fetal heart development, providing new insights into heart development that are crucial for understanding pathological conditions at birth and throughout life.
Collapse
Affiliation(s)
- Reza Amanollahi
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| | - Stacey L. Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| | - Melanie R. Bertossa
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| | - Ashley S. Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| | - Kent L. Thornburg
- Department of Medicine, Center for Developmental Health, Knight Cardiovascular Institute, Bob and Charlee Moore Institute of Nutrition and Wellness, Oregon Health & Science University, Portland, OR 97239, USA;
| | - I. Caroline McMillen
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| | - Michael D. Wiese
- Centre for Pharmaceutical Innovation, Clinical & Health Sciences University of South Australia, Adelaide, SA 5001, Australia;
| | - Mitchell C. Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (M.R.B.); (A.S.M.); (I.C.M.)
| |
Collapse
|
5
|
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
|
6
|
Mishra P, Biesiada I, Gupta P, Ghavami S, Markowski J, Łos MJ. Unraveling the Complexity and Advancements of Transdifferentiation Technologies in the Biomedical Field and Their Potential Clinical Relevance. Arch Immunol Ther Exp (Warsz) 2025; 73:aite-2025-0001. [PMID: 39637369 DOI: 10.2478/aite-2025-0001] [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: 09/11/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Chronic diseases such as cancer, autoimmunity, and organ failure currently depend on conventional pharmaceutical treatment, which may cause detrimental side effects in the long term. In this regard, cell-based therapy has emerged as a suitable alternative for treating these chronic diseases. Transdifferentiation technologies have evolved as a suitable therapeutic alternative that converts one differentiated somatic cell into another phenotype by using transcription factors (TFs), small molecules, or small, single-stranded, non-coding RNA molecules (miRNA). The transdifferentiation techniques rely on simple, fast, standardized, and versatile protocols with minimal chance of tumorigenicity and genotoxicity. However, there are still challenges and limitations that need to be addressed to enhance their clinical translation percentage in the near future. Taking this into account, we have delineated the features and strategies used in the transdifferentiation techniques. Then, we delved into different intermediate states that were attained during transdifferentiation. Advancements in transdifferentiation techniques in the field of tissue engineering, autoimmunity, and cancer therapy were dissected. Furthermore, limitations, challenges, and future perspectives are outlined in this review to provide a whole new picture of the transdifferentiation techniques. Advancements in molecular biology, interdisciplinary research, bioinformatics, and artificial intelligence will push the frontiers of this technology further to establish new avenues for biomedical research.
Collapse
Affiliation(s)
- Purusottam Mishra
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
| | - Izabella Biesiada
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
| | - Payal Gupta
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, India
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Canada
- Faculty of Medicine in Zabrze, University of Technology in Katowice, Zabrze, Poland
| | - Jarosław Markowski
- Department of Laryngology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
- Department of Pathology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
7
|
Carroll A, Bell MJ, Bleach ECL, Turner D, Williams LK. Impact of dairy calf management practices on the intestinal tract microbiome pre-weaning. J Med Microbiol 2025; 74. [PMID: 39879083 DOI: 10.1099/jmm.0.001957] [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] [Indexed: 01/31/2025] Open
Abstract
Introduction. Microbiota in the gastrointestinal tract (GIT) consisting of the rumen and hindgut (the small intestine, cecum and colon) in dairy calves play a vital role in their growth and development. This review discusses the development of dairy calf intestinal microbiomes with an emphasis on the impact that husbandry and rearing management have on microbiome development, health and growth of pre-weaned dairy calves.Discussion. The diversity and composition of the microbes that colonize the lower GIT (small and large intestine) can have a significant impact on the growth and development of the calf, through influence on nutrient metabolism, immune modulation, resistance or susceptibility to infection, production outputs and behaviour modification in adult life. The colonization of the calf intestinal microbiome dynamically changes from birth, increasing microbial richness and diversity until weaning, where further dynamic and drastic microbiome change occurs. In dairy calves, neonatal microbiome development prior to weaning is influenced by direct and indirect factors, some of which could be considered stressors, such as maternal interaction, environment, diet, husbandry and weaning practices. The specific impact of these can dictate intestinal microbial colonization, with potential lifelong consequences.Conclusion. Evidence suggests the potential detrimental effect that sudden changes and stress may have on calf health and growth due to management and husbandry practices, and the importance of establishing a stable yet diverse intestinal microbiome population at an early age is essential for calf success. The possibility of improving the health of calves through intestinal microbiome modulation and using alternative strategies including probiotic use, faecal microbiota transplantation and novel approaches of microbiome tracking should be considered to support animal health and sustainability of dairy production systems.
Collapse
Affiliation(s)
- Aisling Carroll
- Animal and Agriculture Department, Hartpury University, Gloucester, GL19 3BE, UK
| | - Matt J Bell
- Animal and Agriculture Department, Hartpury University, Gloucester, GL19 3BE, UK
| | - Emma C L Bleach
- Animal Science Research Centre, Harper Adams University, Edgmond, Newport, TF10 8NB, Shropshire, UK
| | - Dann Turner
- University of the West of England, Bristol, Coldharbour Lane, BS16 1QY, UK
| | - Lisa K Williams
- Animal and Agriculture Department, Hartpury University, Gloucester, GL19 3BE, UK
| |
Collapse
|
8
|
Chung HJ, Kim JE, Huh Y, Lee JS, Kim SW, Na K, Kim J, Lee SH, Konishi H, Yeo SG, Yon DK, Kim D, Jung J, Jeong NY. The Multi-targeted Effect of Fascaplysin on the Proliferation and Dedifferentiation of Schwann Cells Inhibits Peripheral Nerve Degeneration by Blocking CDK4/6 and Androgen Receptor. Exp Neurobiol 2024; 33:266-281. [PMID: 39806941 PMCID: PMC11738473 DOI: 10.5607/en24025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/15/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025] Open
Abstract
Peripheral neurodegenerative diseases induced by irreversible peripheral nerve degeneration (PND), such as diabetic peripheral neuropathy, have a high prevalence worldwide and reduce the quality of life. However, there is no agent effective against the irreversible PND. After peripheral nerve injury, Schwann cells play an important role in regulating PND. However, because PND involves multiple biochemical events in Schwann cells, a one-drug-single-target therapeutic strategy is not feasible for PND. Here, we suggested that fascaplysin (Fas), a compound with multiple targets (CDK4/6), could overcome these problems. Fas exerted a significant inhibitory effect on axonal degradation, demyelination, and Schwann cell proliferation and dedifferentiation during in vitro and ex vivo PND. To discover the most likely novel target for PND, a chemo-bioinformatics analysis predicted the other on-targets of Fas and identified androgen receptor (AR) which were involved in Schwann cell differentiation and proliferation. AR interacted with Fas, and nuclear import of the AR/Fas complex was inhibited in Schwann cells, altering the expression patterns of transcription factors during PND. Therefore, Fas may have therapeutic potential for irreversible peripheral neurodegenerative diseases.
Collapse
Affiliation(s)
- Hyung-Joo Chung
- Department of Anesthesiology and Pain Medicine, Kosin University, College of Medicine, Busan 49267, Korea
| | - Ja-Eun Kim
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Pharmacology, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Youngbuhm Huh
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Jin San Lee
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Neurology, Kyung Hee University Hospital, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - So-Woon Kim
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Pathology, Kyung Hee University Hospital, College of Medicine, Kyung Hee University, Seoul 02447, Korea, Korea
| | - Kiyong Na
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Pathology, Kyung Hee University Hospital, College of Medicine, Kyung Hee University, Seoul 02447, Korea, Korea
| | - Jiwon Kim
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Seung Hyeun Lee
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Hiroyuki Konishi
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University, Graduate School of Medicine, Yamaguchi 755-8505, Japan
| | - Seung Geun Yeo
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Dong Keon Yon
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Digital Health, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Dokyoung Kim
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Junyang Jung
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Na Young Jeong
- Department of Anatomy and Cell Biology, Dong-A University, College of Medicine, Busan 49201, Korea
| |
Collapse
|
9
|
Ye M, Yang J, Cai Z, Wu J, Xiong W, Hou L. The effect of cortisone on female zebrafish (Dania rerio): Reducing reproductive capacity and offspring survival rate. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 277:107132. [PMID: 39515241 DOI: 10.1016/j.aquatox.2024.107132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
Cortisone is a naturally occurring corticosteroid hormone known for its wide range of anti-inflammatory and immunosuppressive effects, and it is commonly found in various aquatic environments. Previous reports have shown that cortisone can have significant negative impacts on fish; however, its specific effects on fish reproduction have not been thoroughly investigated. In this study, female adult zebrafish were exposed to 0.0 (control), 3.9, 40.2, and 377.9 ng/L of cortisone for 60 days, and multiple endpoints were evaluated. The results showed that as the concentration of cortisone increased, there was an increase in the percentage of perinuclear oocytes and a decrease in the proportion of late-stage oocytes, indicating a stagnation in oocyte development. Additionally, female zebrafish exposed to cortisone exhibited decreased attraction to males and reduced mating intimacy. Furthermore, exposure to cortisone resulted in changes in the development and behavior of zebrafish embryos. At cortisone concentrations of 3.9 and 40.2 ng/L, fewer eggs were laid and the survival rate of fertilized eggs decreased. These observed effects are associated with abnormal transcription levels of genes (Star, Cyp11a1, Cyp17, Cyp19a, Cyp11b, Hsd11β2, Hsd17β3) related to the HPG axis. These findings provided new insights into understanding potential environmental risks associated with corticosteroids.
Collapse
Affiliation(s)
- MeiXin Ye
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - JinLin Yang
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - ZiPing Cai
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - JunHao Wu
- Guangdong Ocean University, Zhanjiang, 524088, China
| | - Wenting Xiong
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China.
| | - LiPing Hou
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China; Key Laboratory of Conservation and Application in Biodiversity of South China, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
10
|
Dong F, Hardy B, Liu J, Mohoric T, Guo W, Exner T, Tong W, Dohler J, Bachler D, Hong H. Development of a comprehensive open access "molecules with androgenic activity resource (MAAR)" to facilitate risk assessment of chemicals. Exp Biol Med (Maywood) 2024; 249:10279. [PMID: 39364092 PMCID: PMC11446862 DOI: 10.3389/ebm.2024.10279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/27/2024] [Indexed: 10/05/2024] Open
Abstract
The increasing prevalence of endocrine-disrupting chemicals (EDCs) and their potential adverse effects on human health underscore the necessity for robust tools to assess and manage associated risks. The androgen receptor (AR) is a critical component of the endocrine system, playing a pivotal role in mediating the biological effects of androgens, which are male sex hormones. Exposure to androgen-disrupting chemicals during critical periods of development, such as fetal development or puberty, may result in adverse effects on reproductive health, including altered sexual differentiation, impaired fertility, and an increased risk of reproductive disorders. Therefore, androgenic activity data is critical for chemical risk assessment. A large amount of androgenic data has been generated using various experimental protocols. Moreover, the data are reported in different formats and in diverse sources. To facilitate utilization of androgenic activity data in chemical risk assessment, the Molecules with Androgenic Activity Resource (MAAR) was developed. MAAR is the first open-access platform designed to streamline and enhance the risk assessment of chemicals with androgenic activity. MAAR's development involved the integration of diverse data sources, including data from public databases and mining literature, to establish a reliable and versatile repository. The platform employs a user-friendly interface, enabling efficient navigation and extraction of pertinent information. MAAR is poised to advance chemical risk assessment by offering unprecedented access to information crucial for evaluating the androgenic potential of a wide array of chemicals. The open-access nature of MAAR promotes transparency and collaboration, fostering a collective effort to address the challenges posed by androgenic EDCs.
Collapse
Affiliation(s)
- Fan Dong
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| | - Barry Hardy
- Edelweiss Connect Inc., Durham, NC, United States
| | - Jie Liu
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| | | | - Wenjing Guo
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| | - Thomas Exner
- Edelweiss Connect Inc., Durham, NC, United States
| | - Weida Tong
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| | - Joh Dohler
- Edelweiss Connect Inc., Durham, NC, United States
| | | | - Huixiao Hong
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| |
Collapse
|
11
|
Le Menuet D, Charalampopoulos IN, Cunningham RL, Kalafatakis K, Nalvarte I. Editorial: Steroid receptors in neuron and glia. Front Endocrinol (Lausanne) 2024; 15:1472908. [PMID: 39205689 PMCID: PMC11349705 DOI: 10.3389/fendo.2024.1472908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Affiliation(s)
- Damien Le Menuet
- INSERM UMRS 1124 (T3S), Faculty of Basic and Biomedical Sciences, Université Paris Cité, Paris, France
| | - Ioannis N. Charalampopoulos
- Division of Basic Sciences, School of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology Hellas (FORTH), Crete, Greece
| | - Rebecca L. Cunningham
- Department of Pharmaceutical Sciences, System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Konstantinos Kalafatakis
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- School of Medicine, University of Crete, Heraklion, Greece
| | - Ivan Nalvarte
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
12
|
Zhong M, Xu W, Tian P, Zhang Q, Wang Z, Liang L, Zhang Q, Yang Y, Lu Y, Wei G. An Inherited Allele Confers Prostate Cancer Progression and Drug Resistance via RFX6/HOXA10-Orchestrated TGFβ Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401492. [PMID: 38932472 PMCID: PMC11348203 DOI: 10.1002/advs.202401492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/01/2024] [Indexed: 06/28/2024]
Abstract
Genetic and epigenetic alterations are cancer hallmark characteristics. However, the role of inherited cancer predisposition alleles in co-opting lineage factor epigenetic reprogramming and tumor progression remains elusive. Here the FinnGen cohort phenome-wide analysis, along with multiple genome-wide association studies, has consistently identified the rs339331-RFX6/6q22 locus associated with prostate cancer (PCa) risk across diverse populations. It is uncovered that rs339331 resides in a reprogrammed androgen receptor (AR) binding site in PCa tumors, with the T risk allele enhancing AR chromatin occupancy. RFX6, an AR-regulated gene linked to rs339331, exhibits synergistic prognostic value for PCa recurrence and metastasis. This comprehensive in vitro and in vivo studies demonstrate the oncogenic functions of RFX6 in promoting PCa cell proliferation and metastasis. Mechanistically, RFX6 upregulates HOXA10 that profoundly correlates with adverse PCa outcomes and is pivotal in RFX6-mediated PCa progression, facilitating the epithelial-mesenchymal transition (EMT) and modulating the TGFβ/SMAD signaling axis. Clinically, HOXA10 elevation is associated with increased EMT scores, tumor advancement and PCa recurrence. Remarkably, reducing RFX6 expression restores enzalutamide sensitivity in resistant PCa cells and tumors. This findings reveal a complex interplay of genetic and epigenetic mechanisms in PCa pathogenesis and drug resistance, centered around disrupted prostate lineage AR signaling and abnormal RFX6 expression.
Collapse
Affiliation(s)
- Mengjie Zhong
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Wenjie Xu
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Pan Tian
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Qin Zhang
- Disease Networks Research UnitFaculty of Biochemistry and Molecular MedicineBiocenter OuluUniversity of OuluOulu90220Finland
| | - Zixian Wang
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Limiao Liang
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Qixiang Zhang
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Yuehong Yang
- Disease Networks Research UnitFaculty of Biochemistry and Molecular MedicineBiocenter OuluUniversity of OuluOulu90220Finland
| | - Ying Lu
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
| | - Gong‐Hong Wei
- MOE Key Laboratory of Metabolism and Molecular Medicine & Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer CenterCancer Institutes, Department of OncologyShanghai Medical College of Fudan UniversityShanghai200032China
- Disease Networks Research UnitFaculty of Biochemistry and Molecular MedicineBiocenter OuluUniversity of OuluOulu90220Finland
| |
Collapse
|
13
|
Díaz-Salazar AJ, Espinosa-Roa A, Saldívar-Guerra E, Pérez-Isidoro R. The disordering effect of SARMs on a biomembrane model. Phys Chem Chem Phys 2024. [PMID: 39040033 DOI: 10.1039/d4cp01002g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
From medicine to sport, selective androgen receptor modulators (SARMs) have represented promising applications. The ability of SARMs to selectively interact with the androgen receptor (AR) indicates that this kind of molecule can interfere with numerous physiological and pathological processes controlled by the AR regulatory mechanism. However, critical concerns in relation to safety and potential side effects of SARMs remain under discussion and investigation. SARMs, being hydrophobic/organic compounds, can be subjected to hydrophobic interactions. In this perspective, we hypothesize that SARMs interact with lipid membranes, producing significant physical and chemical changes that could be associated with several effects that SARMs represent in biological systems. In this context, the effect of SARMs on lipid membranes mediated by non-specific interactions is little explored. Here, we report significant information related to the changes that ostarine, ligandrol, andarine, and cardarine produce in the thermodynamic properties of a lipid biomembrane model. Physical changes and chemical interactions of the systems were investigated by differential scanning calorimetry (DSC), dynamic light scattering (DLS), attenuated total reflectance-Fourier transform infrared spectroscopy (ATR-FTIR), and theoretical calculations implementing density functional theory (DFT). We demonstrate that ostarine, ligandrol, andarine, and cardarine can strongly interact with a lipid biomembrane model composed of 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC), and accordingly, these molecules can be incorporated into the polar/hydrophobic regions of the lipid bilayer. By employing theoretical calculations, we gained insights into the possible electrostatic interactions between SARMs and phospholipid molecules, enhancing our understanding of the driving forces behind the interactions of SARMs with lipid membranes. Overall, this investigation provides relevant knowledge related to the biophysical-chemical effects that SARMs produce in biomembrane models and could be of practical reference for promising applications of SARMs in medicine and sport.
Collapse
Affiliation(s)
- Alma Jessica Díaz-Salazar
- Laboratorio de Bio-fisicoquímica. Departamento de Fisicoquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico.
| | - Arián Espinosa-Roa
- CONAHCyT-Centro de Investigación en Química Aplicada (CIQA), Unidad Monterrey. Alianza Sur no. 204, Parque de Investigación en Innovación Tecnológica (PIIT), km 10 autopista internacional Mariano Escobedo, C.P. 66628, Apodaca, Nuevo León, Mexico.
| | - Enrique Saldívar-Guerra
- Centro de Investigación en Química Aplicada (CIQA), Enrique Reyna, 140, 25294 Saltillo Coahuila, Mexico.
| | - Rosendo Pérez-Isidoro
- Centro de Investigación en Química Aplicada (CIQA), Enrique Reyna, 140, 25294 Saltillo Coahuila, Mexico.
| |
Collapse
|
14
|
Toso A, Garoche C, Balaguer P. Human and fish differences in steroid receptors activation: A review. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 948:174889. [PMID: 39047839 DOI: 10.1016/j.scitotenv.2024.174889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/05/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Steroid receptors (SRs) are transcription factors activated by steroid hormones (SHs) that belong to the nuclear receptors (NRs) superfamily. Several studies have shown that SRs are targets of endocrine disrupting chemicals (EDCs), widespread substances in the environment capable of interfering with the endogenous hormonal pathways and causing adverse health effects in living organisms and/or their progeny. Cell lines with SRs reporter gene are currently used for in vitro screening of large quantities of chemicals with suspected endocrine-disrupting activities. However, most of these cell lines express human SRs and therefore the toxicological data obtained are also extrapolated to non-mammalian species. In parallel, in vivo tests have recently been developed on fish species whose data are also extrapolated to mammalian species. As some species-specific differences in SRs activation by natural and synthetic chemicals have been recently reported, the aim of this review is to summarize those between human and fish SRs, as representatives of mammalian and non-mammalian toxicology, respectively. Overall, this literature study aims to improve inter-species extrapolation of toxicological data on EDCs and to understand which reporter gene cell lines expressing human SRs are relevant for the assessment of effects in fish and whether in vivo tests on fish can be properly used in the assessment of adverse effects on human health.
Collapse
Affiliation(s)
- Anna Toso
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut régional du Cancer de Montpellier (ICM), 34090 Montpellier, France; Department Environmental Toxicology, Swiss Federal Institute of Aquatic Science and Technology, Eawag, 8600 Dübendorf, Switzerland.
| | - Clémentine Garoche
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut régional du Cancer de Montpellier (ICM), 34090 Montpellier, France
| | - Patrick Balaguer
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut régional du Cancer de Montpellier (ICM), 34090 Montpellier, France
| |
Collapse
|
15
|
Lockett J, Inder WJ, Clifton VL. The Glucocorticoid Receptor: Isoforms, Functions, and Contribution to Glucocorticoid Sensitivity. Endocr Rev 2024; 45:593-624. [PMID: 38551091 PMCID: PMC11244253 DOI: 10.1210/endrev/bnae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Indexed: 07/13/2024]
Abstract
Glucocorticoids exert pleiotropic effects on all tissues to regulate cellular and metabolic homeostasis. Synthetic forms are used therapeutically in a wide range of conditions for their anti-inflammatory benefits, at the cost of dose and duration-dependent side effects. Significant variability occurs between tissues, disease states, and individuals with regard to both the beneficial and deleterious effects. The glucocorticoid receptor (GR) is the site of action for these hormones and a vast body of work has been conducted understanding its function. Traditionally, it was thought that the anti-inflammatory benefits of glucocorticoids were mediated by transrepression of pro-inflammatory transcription factors, while the adverse metabolic effects resulted from direct transactivation. This canonical understanding of the GR function has been brought into question over the past 2 decades with advances in the resolution of scientific techniques, and the discovery of multiple isoforms of the receptor present in most tissues. Here we review the structure and function of the GR, the nature of the receptor isoforms, and the contribution of the receptor to glucocorticoid sensitivity, or resistance in health and disease.
Collapse
Affiliation(s)
- Jack Lockett
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4101, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Metro South Health, Woolloongabba, QLD 4102, Australia
| | - Warrick J Inder
- Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Metro South Health, Woolloongabba, QLD 4102, Australia
| | - Vicki L Clifton
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4101, Australia
| |
Collapse
|
16
|
Ochiai K, Yonezawa R, Fujii S. Structural Development of Androgen Receptor Antagonists Using Phenylferrocene Framework as a Hydrophobic Pharmacophore. ChemMedChem 2024; 19:e202400040. [PMID: 38291942 DOI: 10.1002/cmdc.202400040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 02/01/2024]
Abstract
We previously identified nitrophenylferrocenes and cyanophenylferrocenes as promising lead structures of novel androgen receptor (AR) antagonists, based on the structural similarity between ferrocene and the steroidal skeleton. In the present research, we explored the structure-activity relationship (SAR) of phenylferrocene derivatives. Introduction of a hydrophobic substituent such as a chlorine atom at the 2-position or 3-position of phenylferrocene derivatives significantly increased the antagonistic activity toward wild-type AR, and among the synthesized compounds, 3-chloro-4-cyanophenylferrocene (29) exhibited the most potent anti-proliferative activity toward the androgen-dependent growth of SC-3 cells expressing wild-type AR (IC50 14 nM). Like conventional antiandrogens such as hydroxyflutamide, the major active metabolite of flutamide, compound 29 exhibited agonistic activity toward T877A-AR, a mutant AR expressed in human prostate cancer cell line LNCaP. Notably, however, the 2-chloro isomer 27 showed potent antagonistic activity toward wild-type AR (IC50 49 nM) and also exhibited antagonistic activity toward T877A-AR. Our SAR data should prove helpful for the development of new-generation AR antagonists based on phenylferrocene as candidate agents to treat drug-resistant prostate cancer.
Collapse
Affiliation(s)
- Kotaro Ochiai
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Ryo Yonezawa
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Shinya Fujii
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| |
Collapse
|
17
|
Gao H, Zhang JY, Zhao LJ, Guo YY. Synthesis and application of clinically approved small-molecule drugs targeting androgen receptor. Bioorg Chem 2024; 143:106998. [PMID: 38035513 DOI: 10.1016/j.bioorg.2023.106998] [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: 09/10/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023]
Abstract
Androgen receptor (AR) plays a crucial role in various physiological processes. Dysregulation of AR signaling has been implicated in several diseases, such as prostate cancer and androgenetic alopecia. Therefore, the development of drugs that specifically target AR has gained significant attention in the field of drug discovery. This review provides an overview of the synthetic routes of clinically approved small molecule drugs targeting AR and discusses the clinical applications of these drugs in the treatment of AR-related diseases. The review also highlights the challenges and future perspectives in this field, including the need for improved drug design and the exploration of novel therapeutic targets. Through an integrated analysis of the therapeutic applications, synthetic methodologies, and mechanisms of action associated with these approved drugs, this review facilitates a holistic understanding of the versatile roles and therapeutic potential of AR-targeted interventions. Overall, this comprehensive review serves as a valuable resource for medicinal chemists interested in the development of small-molecule drugs targeting AR.
Collapse
Affiliation(s)
- Hua Gao
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jing-Yi Zhang
- The Rogel Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States; College of Chemistry and Chemical Engineering, Zhengzhou Normal University, 450044, China.
| | - Li-Jie Zhao
- The Rogel Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Yuan-Yuan Guo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
18
|
Mehralivand S, Thomas C, Puhr M, Claessens F, van de Merbel AF, Dubrovska A, Jenster G, Bernemann C, Sommer U, Erb HHH. New advances of the androgen receptor in prostate cancer: report from the 1st International Androgen Receptor Symposium. J Transl Med 2024; 22:71. [PMID: 38238739 PMCID: PMC10795409 DOI: 10.1186/s12967-024-04878-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 01/10/2024] [Indexed: 01/22/2024] Open
Abstract
The androgen receptor (AR) is a crucial player in various aspects of male reproduction and has been associated with the development and progression of prostate cancer (PCa). Therefore, the protein is the linchpin of current PCa therapies. Despite great research efforts, the AR signaling pathway has still not been deciphered, and the emergence of resistance is still the biggest problem in PCa treatment. To discuss the latest developments in AR research, the "1st International Androgen Receptor Symposium" offered a forum for the exchange of clinical and scientific innovations around the role of the AR in prostate cancer (PCa) and to stimulate new collaborative interactions among leading scientists from basic, translational, and clinical research. The symposium included three sessions covering preclinical studies, prognostic and diagnostic biomarkers, and ongoing prostate cancer clinical trials. In addition, a panel discussion about the future direction of androgen deprivation therapy and anti-AR therapy in PCa was conducted. Therefore, the newest insights and developments in therapeutic strategies and biomarkers are discussed in this report.
Collapse
Affiliation(s)
- Sherif Mehralivand
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Christian Thomas
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Martin Puhr
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | | | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Institute of Radiooncology-OncoRay, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Guido Jenster
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | | | - Ulrich Sommer
- Institut für Pathologie, Medical Faculty, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Holger H H Erb
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
19
|
Stölting G, Scholl UI. Adrenal Anion Channels: New Roles in Zona Glomerulosa Physiology and in the Pathophysiology of Primary Aldosteronism. Handb Exp Pharmacol 2024; 283:59-79. [PMID: 37495852 DOI: 10.1007/164_2023_680] [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] [Indexed: 07/28/2023]
Abstract
The mineralocorticoid aldosterone is produced in the zona glomerulosa of the adrenal cortex. Its synthesis is regulated by the serum concentrations of the peptide hormone angiotensin II and potassium. The primary role of aldosterone is to control blood volume and electrolytes. The autonomous production of aldosterone (primary aldosteronism, PA) is considered the most frequent cause of secondary hypertension. Aldosterone-producing adenomas and (micro-)nodules are frequent causes of PA and often carry somatic mutations in ion channels and transporters. Rare familial forms of PA are due to germline mutations. Both somatic and germline mutations in the chloride channel gene CLCN2, encoding ClC-2, have been identified in PA. Clinical findings and results from cell culture and animal models have advanced our knowledge about the role of anions in PA. The zona glomerulosa of the adrenal gland has now been firmly established as a tissue in which anions play a significant role for signaling. In this overview, we aim to summarize the current knowledge and highlight novel concepts as well as open questions.
Collapse
Affiliation(s)
- Gabriel Stölting
- Center of Functional Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ute I Scholl
- Center of Functional Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
20
|
Patel VK, Vaishnaw A, Shirbhate E, Kore R, Singh V, Veerasamy R, Rajak H. Cortisol as a Target for Treating Mental Disorders: A Promising Avenue for Therapy. Mini Rev Med Chem 2024; 24:588-600. [PMID: 37861053 DOI: 10.2174/0113895575262104230928042150] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/19/2023] [Accepted: 08/05/2023] [Indexed: 10/21/2023]
Abstract
Cortisol, commonly known as the "stress hormone," plays a critical role in the body's response to stress. Elevated cortisol levels have been associated with various mental disorders, including anxiety, depression, and post-traumatic stress disorder. Consequently, researchers have explored cortisol modulation as a promising avenue for treating these conditions. However, the availability of research on cortisol as a therapeutic option for mental disorders is limited, and existing studies employ diverse methodologies and outcome measures. This review article aimed to provide insights into different treatment approaches, both pharmacological and non-pharmacological, which can effectively modulate cortisol levels. Pharmacological interventions involve the use of substances, such as somatostatin analogs, dopamine agonists, corticotropin-releasing hormone antagonists, and cortisol synthesis inhibitors. Additionally, non-pharmacological techniques, including cognitivebehavioral therapy, herbs and supplements, transcranial magnetic stimulation, lifestyle changes, and surgery, have been investigated to reduce cortisol levels. The emerging evidence suggests that cortisol modulation could be a promising treatment option for mental disorders. However, more research is needed to fully understand the effectiveness and safety of these therapies.
Collapse
Affiliation(s)
- Vijay K Patel
- Pushpendra College of Pharmacy, Ambikapur, Surguja 497101, (C.G.), India
| | - Aayush Vaishnaw
- Dr. C.V. Raman Institute of Pharmacy, Dr. C.V. Raman University, Bilaspur, C.G. 495113, India
| | - Ekta Shirbhate
- Department of Pharmacy, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| | - Rakesh Kore
- Department of Pharmacy, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| | - Vaibhav Singh
- Department of Pharmacy, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| | - Ravichandran Veerasamy
- Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Harish Rajak
- Department of Pharmacy, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| |
Collapse
|
21
|
Asemota S, Effah W, Young KL, Holt J, Cripe L, Ponnusamy S, Thiyagarajan T, Hwang DJ, He Y, Mcnamara K, Johnson D, Wang Y, Grimes B, Khosrosereshki Y, Hollingsworth TJ, Fleming MD, Pritchard FE, Hendrix A, Khan F, Fan M, Makowski L, Yin Z, Sasano H, Hayes DN, Pfeffer LM, Miller DD, Narayanan R. Identification of a targetable JAK-STAT enriched androgen receptor and androgen receptor splice variant positive triple-negative breast cancer subtype. Cell Rep 2023; 42:113461. [PMID: 37979170 PMCID: PMC10872270 DOI: 10.1016/j.celrep.2023.113461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/18/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype with no targeted therapeutics. The luminal androgen receptor (LAR) subtype constitutes 15% of TNBC and is enriched for androgen receptor (AR) and AR target genes. Here, we show that a cohort of TNBC not only expresses AR at a much higher rate (∼80%) but also expresses AR splice variants (AR-SVs) (∼20%), further subclassifying LAR-TNBC. Higher AR and AR-SV expression and corresponding aggressive phenotypes are observed predominantly in specimens obtained from African American women. LAR TNBC specimens are enriched for interferon, Janus kinase (JAK)-signal activator and transducer (STAT), and androgen signaling pathways, which are exclusive to AR-expressing epithelial cancer cells. AR- and AR-SV-expressing TNBC cell proliferation and xenograft and patient-tumor explant growth are inhibited by AR N-terminal domain-binding selective AR degrader or by a JAK inhibitor. Biochemical analysis suggests that STAT1 is an AR coactivator. Collectively, our work identifies pharmacologically targetable TNBC subtypes and identifies growth-promoting interaction between AR and JAK-STAT signaling.
Collapse
Affiliation(s)
- Sarah Asemota
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Wendy Effah
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Kirsten L Young
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Jeremiah Holt
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Linnea Cripe
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Suriyan Ponnusamy
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Thirumagal Thiyagarajan
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Yali He
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Keely Mcnamara
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8577, Japan
| | - Daniel Johnson
- Molecular Bioinformatics Core, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Yinan Wang
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Brandy Grimes
- West Cancer Center and Research Institute, Memphis, TN 38138, USA
| | - Yekta Khosrosereshki
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - T J Hollingsworth
- Department of Ophthalmology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Martin D Fleming
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Frances E Pritchard
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Ashley Hendrix
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Farhan Khan
- Department of Pathology, Methodist Hospital, Memphis, TN 38104, USA
| | - Meiyun Fan
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Liza Makowski
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA; UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Zheng Yin
- Biomedical and Informatics Services Core, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8577, Japan
| | - D Neil Hayes
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA; UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Lawrence M Pfeffer
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA; UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38103, USA; UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Ramesh Narayanan
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA; UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| |
Collapse
|
22
|
Chen Y, Fei X, Liu G, Li X, Huang L, Yang LZ, Li Y, Xu B, Fang W. P-Glycoprotein Exacerbates Brain Injury Following Experimental Cerebral Ischemia by Promoting Proinflammatory Microglia Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:6916819. [PMID: 38144707 PMCID: PMC10748718 DOI: 10.1155/2023/6916819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/02/2023] [Accepted: 11/24/2023] [Indexed: 12/26/2023]
Abstract
Microglia are activated following cerebral ischemic insult. P-glycoprotein (P-gp) is an efflux transporter on microvascular endothelial cells and upregulated after cerebral ischemia. This study evaluated the effects and possible mechanisms of P-gp on microglial polarization/activation in mice after ischemic stroke. P-gp-specific siRNA and adeno-associated virus (p-AAV) were used to silence and overexpress P-gp, respectively. Middle cerebral artery occlusion/reperfusion (MCAO/R) and oxygen-glucose deprivation/reoxygenation (OGD/R) were performed in mice and cerebral microvascular endothelial cells (bEnd.3) in vitro, respectively. OGD/R-injured bEnd.3 cells were cocultured with mouse microglial cells (BV2) in Transwell. Influences on acute ischemic stroke outcome, the expression of inflammatory cytokines, and chemokines and chemokines receptors, microglial polarization, glucocorticoid receptor (GR) nuclear translocation, and GR-mediated mRNA decay (GMD) activation were evaluated via reverse transcription real-time polymerase chain reaction, western blot, or immunofluorescence. Silencing P-gp markedly alleviated experimental ischemia injury as indicated by reduced cerebral infarct size, improved neurological deficits, and reduced the expression of interleukin-6 (IL-6) and IL-12 expression. Silencing P-gp also mitigated proinflammatory microglial polarization and the expression of C-C motif chemokine ligand 2 (CCL2) and its receptor CCR2 expression, whereas promoted anti-inflammatory microglia polarization. Additionally, P-gp silencing promoted GR nuclear translocation and the expression of GMD relative proteins in endothelial cells. Conversely, overexpressing P-gp via p-AAV transfection offset all these effects. Furthermore, silencing endothelial GR counteracted all effects mediated by silencing or overexpressing P-gp. Elevated P-gp expression aggravated inflammatory response and brain damage after ischemic stroke by augmenting proinflammatory microglial polarization in association with increased endothelial CCL2 release due to GMD inhibition by P-gp.
Collapse
Affiliation(s)
- Yan Chen
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Xuan Fei
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Ge Liu
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Xiang Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Liangliang Huang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Lele Zixin Yang
- Penn State University, University Park, State College, PA 16802, USA
| | - Yunman Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| |
Collapse
|
23
|
Li S, Ying Z, Gentenaar M, Rensen PCN, Kooijman S, Visser JA, Meijer OC, Kroon J. Glucocorticoid Receptor Antagonism Improves Glucose Metabolism in a Mouse Model of Polycystic Ovary Syndrome. J Endocr Soc 2023; 8:bvad162. [PMID: 38169733 PMCID: PMC10758754 DOI: 10.1210/jendso/bvad162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Indexed: 01/05/2024] Open
Abstract
Context Polycystic ovary syndrome (PCOS) is a complex metabolic disorder associated with obesity, insulin resistance, and dyslipidemia. Hyperandrogenism is a major characteristic of PCOS. Increased androgen exposure is believed to deregulate metabolic processes in various tissues as part of the PCOS pathogenesis, predominantly through the androgen receptor (AR). Notably, various metabolic features in PCOS are similar to those observed after excess glucocorticoid exposure. Objective We hypothesized that glucocorticoid receptor (GR) signaling is involved in the metabolic symptoms of PCOS. Methods In a PCOS model of chronic dihydrotestosterone (DHT) exposure in female mice, we investigated whether GR signaling machinery was (de)regulated, and if treatment with a selective GR antagonist alleviated the metabolic symptoms. Results We observed an upregulation of GR messenger RNA expression in the liver after DHT exposure. In white adipose tissues and liver we found that DHT upregulated Hsd11b1, which encodes for the enzyme that converts inactive into active glucocorticoids. We found that preventive but not therapeutic administration of a GR antagonist alleviated DHT-induced hyperglycemia and restored glucose tolerance. We did not observe strong effects of GR antagonism in DHT-exposed mice on other features like total fat mass and lipid accumulation in various tissues. Conclusion We conclude that GR activation may play a role in glucose metabolism in DHT-exposed mice.
Collapse
Affiliation(s)
- Sheng Li
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Zhixiong Ying
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Max Gentenaar
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Jenny A Visser
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, the Netherlands
| | - Onno C Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Jan Kroon
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| |
Collapse
|
24
|
Kraemer RR, Kraemer BR. The effects of peripheral hormone responses to exercise on adult hippocampal neurogenesis. Front Endocrinol (Lausanne) 2023; 14:1202349. [PMID: 38084331 PMCID: PMC10710532 DOI: 10.3389/fendo.2023.1202349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023] Open
Abstract
Over the last decade, a considerable amount of new data have revealed the beneficial effects of exercise on hippocampal neurogenesis and the maintenance or improvement of cognitive function. Investigations with animal models, as well as human studies, have yielded novel understanding of the mechanisms through which endocrine signaling can stimulate neurogenesis, as well as the effects of exercise on acute and/or chronic levels of these circulating hormones. Considering the effects of aging on the decline of specific endocrine factors that affect brain health, insights in this area of research are particularly important. In this review, we discuss how different forms of exercise influence the peripheral production of specific endocrine factors, with particular emphasis on brain-derived neurotrophic factor, growth hormone, insulin-like growth factor-1, ghrelin, estrogen, testosterone, irisin, vascular endothelial growth factor, erythropoietin, and cortisol. We also describe mechanisms through which these endocrine responses to exercise induce cellular changes that increase hippocampal neurogenesis and improve cognitive function.
Collapse
Affiliation(s)
- Robert R. Kraemer
- Department of Kinesiology and Health Studies, Southeastern Louisiana University, Hammond, LA, United States
| | - Bradley R. Kraemer
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, AL, United States
| |
Collapse
|
25
|
Murphy CN, Delles C, Davies E, Connelly PJ. Cardiovascular disease in transgender individuals. Atherosclerosis 2023; 384:117282. [PMID: 37821271 DOI: 10.1016/j.atherosclerosis.2023.117282] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/23/2023] [Accepted: 09/05/2023] [Indexed: 10/13/2023]
Abstract
The population of people identifying as transgender has grown rapidly in recent years, resulting in a substantive increase in individuals obtaining gender-affirming medical care to align their secondary sex characteristics with their gender identity. This has established benefits for patients including improvements in gender dysphoria and psychosocial functioning, while reducing adverse mental health outcomes. Despite these potential advantages, recent evidence has suggested that gender-affirming hormone therapy (GAHT) may increase the risk of cardiovascular disease. However, owing to a paucity of research, the mechanisms underpinning these increased risks are poorly understood. Moreover, previous research has been limited by heterogenous methodologies, being underpowered, and lacking appropriate control populations. Consequently, the need for evidence regarding cardiovascular health in LGBTQ + individuals has been recognised as a critical area for future research to facilitate better healthcare and guidance. Recent research investigating the effect of transmasculine (testosterone) GAHT on cardiovascular disease risk points to testosterone effecting the nitric oxide pathway, triggering inflammation, and promoting endothelial dysfunction. Equivalent studies focussing on transfeminine (oestrogen) GAHT are required, representing a crucial area of future research. Furthermore, when examining the effects of GAHT on the vasculature, it cannot be ignored that there are multiple factors that may increase the burden of cardiovascular disease in the transgender population. Such stressors include major psychological stress; increased adverse health behaviours, such as smoking; discrimination; and lowered socioeconomic status; all of which undoubtedly impact upon cardiovascular disease risk and offers the opportunity for intervention.
Collapse
Affiliation(s)
- Charlotte N Murphy
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom
| | - Christian Delles
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom
| | - Eleanor Davies
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom
| | - Paul J Connelly
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom.
| |
Collapse
|
26
|
Onyango DO, Selman BG, Rose JL, Ellison CA, Nash JF. Comparison between endocrine activity assessed using ToxCast/Tox21 database and human plasma concentration of sunscreen active ingredients/UV filters. Toxicol Sci 2023; 196:25-37. [PMID: 37561120 PMCID: PMC10613966 DOI: 10.1093/toxsci/kfad082] [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] [Indexed: 08/11/2023] Open
Abstract
Sunscreen products are composed of ultraviolet (UV) filters and formulated to reduce exposure to sunlight thereby lessening skin damage. Concerns have been raised regarding the toxicity and potential endocrine disrupting (ED) effects of UV filters. The ToxCast/Tox21 program, that is, CompTox, is a high-throughput in vitro screening database of chemicals that identify adverse outcome pathways, key events, and ED potential of chemicals. Using the ToxCast/Tox21 database, octisalate, homosalate, octocrylene, oxybenzone, octinoxate, and avobenzone, 6 commonly used organic UV filters, were found to have been evaluated. These UV filters showed low potency in these bioassays with most activity detected above the range of the cytotoxic burst. The pathways that were most affected were the cell cycle and the nuclear receptor pathways. Most activity was observed in liver and kidney-based bioassays. These organic filters and their metabolites showed relatively weak ED activity when tested in bioassays measuring estrogen receptor (ER), androgen receptor (AR), thyroid receptor, and steroidogenesis activity. Except for oxybenzone, all activity in the endocrine assays occurred at concentrations greater than the cytotoxic burst. Moreover, except for oxybenzone, plasma concentrations (Cmax) measured in humans were at least 100× lower than bioactive (AC50/ACC) concentrations that produced a response in ToxCast/Tox21 assays. These data are consistent with in vivo animal/human studies showing weak or negligible endocrine activity. In sum, when considered as part of a weight-of-evidence assessment and compared with measured plasma concentrations, the results show these organic UV filters have low intrinsic biological activity and risk of toxicity including endocrine disruption in humans.
Collapse
Affiliation(s)
- David O Onyango
- Global Product Stewardship, The Procter & Gamble Company, Mason, Ohio 45040, USA
| | - Bastian G Selman
- Global Product Stewardship, The Procter & Gamble Company, Mason, Ohio 45040, USA
| | - Jane L Rose
- Global Product Stewardship, The Procter & Gamble Company, Mason, Ohio 45040, USA
| | - Corie A Ellison
- Global Product Stewardship, The Procter & Gamble Company, Mason, Ohio 45040, USA
| | - J F Nash
- Global Product Stewardship, The Procter & Gamble Company, Mason, Ohio 45040, USA
| |
Collapse
|
27
|
Ammari R, Monaca F, Cao M, Nassar E, Wai P, Del Grosso NA, Lee M, Borak N, Schneider-Luftman D, Kohl J. Hormone-mediated neural remodeling orchestrates parenting onset during pregnancy. Science 2023; 382:76-81. [PMID: 37797007 PMCID: PMC7615220 DOI: 10.1126/science.adi0576] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/09/2023] [Indexed: 10/07/2023]
Abstract
During pregnancy, physiological adaptations prepare the female body for the challenges of motherhood. Becoming a parent also requires behavioral adaptations. Such adaptations can occur as early as during pregnancy, but how pregnancy hormones remodel parenting circuits to instruct preparatory behavioral changes remains unknown. We found that action of estradiol and progesterone on galanin (Gal)-expressing neurons in the mouse medial preoptic area (MPOA) is critical for pregnancy-induced parental behavior. Whereas estradiol silences MPOAGal neurons and paradoxically increases their excitability, progesterone permanently rewires this circuit node by promoting dendritic spine formation and recruitment of excitatory synaptic inputs. This MPOAGal-specific neural remodeling sparsens population activity in vivo and results in persistently stronger, more selective responses to pup stimuli. Pregnancy hormones thus remodel parenting circuits in anticipation of future behavioral need.
Collapse
Affiliation(s)
- Rachida Ammari
- State-dependent Neural Processing Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Francesco Monaca
- State-dependent Neural Processing Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Mingran Cao
- State-dependent Neural Processing Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Estelle Nassar
- State-dependent Neural Processing Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Patty Wai
- State-dependent Neural Processing Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Nicholas A. Del Grosso
- State-dependent Neural Processing Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Matthew Lee
- State-dependent Neural Processing Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Neven Borak
- State-dependent Neural Processing Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Deborah Schneider-Luftman
- Bioinformatics and Biostatistics Science Technology Platform, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Johannes Kohl
- State-dependent Neural Processing Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| |
Collapse
|
28
|
Wilton-Clark H, Al-aghbari A, Yang J, Yokota T. Advancing Epidemiology and Genetic Approaches for the Treatment of Spinal and Bulbar Muscular Atrophy: Focus on Prevalence in the Indigenous Population of Western Canada. Genes (Basel) 2023; 14:1634. [PMID: 37628685 PMCID: PMC10454234 DOI: 10.3390/genes14081634] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/10/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA), also known as Kennedy's disease, is a debilitating neuromuscular disease characterized by progressive muscular weakness and neuronal degeneration, affecting 1-2 individuals per 100,000 globally. While SBMA is relatively rare, recent studies have shown a significantly higher prevalence of the disease among the indigenous population of Western Canada compared to the general population. The disease is caused by a pathogenic expansion of polyglutamine residues in the androgen receptor protein, which acts as a key transcriptional regulator for numerous genes. SBMA has no cure, and current treatments are primarily supportive and focused on symptom management. Recently, a form of precision medicine known as antisense therapy has gained traction as a promising therapeutic option for numerous neuromuscular diseases. Antisense therapy uses small synthetic oligonucleotides to confer therapeutic benefit by acting on pathogenic mRNA molecules, serving to either degrade pathogenic mRNA transcripts or helping to modulate splicing. Recent studies have explored the suitability of antisense therapy for the treatment of SBMA, primarily focused on gene therapy and antisense-mediated mRNA knockdown approaches. Advancements in understanding the pathogenesis of SBMA and the development of targeted therapies offer hope for improved quality of life for individuals affected by this debilitating condition. Continued research is essential to optimize these genetic approaches, ensuring their safety and efficacy.
Collapse
Affiliation(s)
- Harry Wilton-Clark
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| | - Ammar Al-aghbari
- Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| | - Jessica Yang
- Department of Immunology, Department of Pharmacology and Toxicology, Faculty of Arts and Science, University of Toronto, Toronto, ON M5S 1A1, Canada;
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| |
Collapse
|
29
|
Toso A, Boulahtouf A, Escande A, Garoche C, Balaguer P. A comparative study of human and zebrafish glucocorticoid receptor activities of natural and pharmaceutical steroids. Front Endocrinol (Lausanne) 2023; 14:1235501. [PMID: 37654569 PMCID: PMC10466050 DOI: 10.3389/fendo.2023.1235501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/31/2023] [Indexed: 09/02/2023] Open
Abstract
Introduction The action of environmental steroids on the human glucocorticoid receptor (hGR) has been pointed out with the risk to impair physiological immune and metabolic processes regulated by this nuclear receptor. However, there is still a lack of mechanistic information regarding their ability to interact with GR in aquatic species. Methods To investigate ligand activation differences between hGR and zebrafish GR (zfGR), we tested several natural and synthetic steroids using reporter cell lines expressing hGR or zfGR. Results and discussion Almost all the glucocorticoids tested (dexamethasone, cortisol, bimedrazol, medrol, cortivazol and fluticasone) are agonists of the two receptors with similar potencies. The dissociated glucocorticoids, RU24782 and RU24858 are agonists of both zfGR and hGR but with a better potency for the latter. On the other hand, the synthetic glucocorticoid forbimenol and the mineralocorticoid aldosterone are agonist on hGR but antagonist on zfGR. The other steroids tested, androgens and progestins, are all antagonists of both GRs with equal or lower potency on zfGR than on hGR. Surprisingly, the lower efficacy and potency on zfGR of aldosterone, forbimenol and the dissociated glucocorticoids is not related to their affinity for the receptors which would suggest that it could be related to less efficacious recruitment of coactivators by zfGR compared to hGR.
Collapse
Affiliation(s)
- Anna Toso
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Abdelhay Boulahtouf
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Aurélie Escande
- UMR Hydrosciences Montpellier, Université de Montpellier, Montpellier, France
| | - Clémentine Garoche
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Patrick Balaguer
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| |
Collapse
|
30
|
Bhaumik S, Lockett J, Cuffe J, Clifton VL. Glucocorticoids and Their Receptor Isoforms: Roles in Female Reproduction, Pregnancy, and Foetal Development. BIOLOGY 2023; 12:1104. [PMID: 37626990 PMCID: PMC10452123 DOI: 10.3390/biology12081104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023]
Abstract
Alterations in the hypothalamic-pituitary-adrenal (HPA) axis and associated changes in circulating levels of glucocorticoids are integral to an organism's response to stressful stimuli. Glucocorticoids acting via glucocorticoid receptors (GRs) play a role in fertility, reproduction, placental function, and foetal development. GRs are ubiquitously expressed throughout the female reproductive system and regulate normal reproductive function. Stress-induced glucocorticoids have been shown to inhibit reproduction and affect female gonadal function by suppressing the hypothalamic-pituitary-gonadal (HPG) axis at each level. Furthermore, during pregnancy, a mother's exposure to prenatal stress or external glucocorticoids can result in long-lasting alterations to the foetal HPA and neuroendocrine function. Several GR isoforms generated via alternative splicing or translation initiation from the GR gene have been identified in the mammalian ovary and uterus. The GR isoforms identified include the splice variants, GRα and GRβ, and GRγ and GR-P. Glucocorticoids can exert both stimulatory and inhibitory effects and both pro- and anti-inflammatory functions in the ovary, in vitro. In the placenta, thirteen GR isoforms have been identified in humans, guinea pigs, sheep, rats, and mice, indicating they are conserved across species and may be important in mediating a differential response to stress. Distinctive responses to glucocorticoids, differential birth outcomes in pregnancy complications, and sex-based variations in the response to stress could all potentially be dependent on a particular GR expression pattern. This comprehensive review provides an overview of the structure and function of the GR in relation to female fertility and reproduction and discusses the changes in the GR and glucocorticoid signalling during pregnancy. To generate this overview, an extensive non-systematic literature search was conducted across multiple databases, including PubMed, Web of Science, and Google Scholar, with a focus on original research articles, meta-analyses, and previous review papers addressing the subject. This review integrates the current understanding of GR variants and their roles in glucocorticoid signalling, reproduction, placental function, and foetal growth.
Collapse
Affiliation(s)
- Sreeparna Bhaumik
- Mater Research Institute, Faculty of Medicine, The University of Queensland, Brisbane 4067, Australia; (S.B.); (J.L.)
| | - Jack Lockett
- Mater Research Institute, Faculty of Medicine, The University of Queensland, Brisbane 4067, Australia; (S.B.); (J.L.)
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Metro South Health, Brisbane 4102, Australia
| | - James Cuffe
- School of Biomedical Sciences, The University of Queensland, Brisbane 4067, Australia;
| | - Vicki L. Clifton
- Mater Research Institute, Faculty of Medicine, The University of Queensland, Brisbane 4067, Australia; (S.B.); (J.L.)
| |
Collapse
|
31
|
Alatkar SA, Wang D. CMOT: Cross-Modality Optimal Transport for multimodal inference. Genome Biol 2023; 24:163. [PMID: 37434182 PMCID: PMC10334579 DOI: 10.1186/s13059-023-02989-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 06/14/2023] [Indexed: 07/13/2023] Open
Abstract
Multimodal measurements of single-cell sequencing technologies facilitate a comprehensive understanding of specific cellular and molecular mechanisms. However, simultaneous profiling of multiple modalities of single cells is challenging, and data integration remains elusive due to missing modalities and cell-cell correspondences. To address this, we developed a computational approach, Cross-Modality Optimal Transport (CMOT), which aligns cells within available multi-modal data (source) onto a common latent space and infers missing modalities for cells from another modality (target) of mapped source cells. CMOT outperforms existing methods in various applications from developing brain, cancers to immunology, and provides biological interpretations improving cell-type or cancer classifications.
Collapse
Affiliation(s)
- Sayali Anil Alatkar
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI, 53076, USA
| | - Daifeng Wang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI, 53076, USA.
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, 53076, USA.
| |
Collapse
|
32
|
Wang X, Chang L, Li Z, Jiang Y, Chen Y, Jia X, Wang Q, Ren X, Ma Z, Zhang W. Newly identified adverse events of enzalutamide using the food and drug administration adverse event reporting system. Expert Opin Drug Saf 2023; 22:1099-1103. [PMID: 37670490 DOI: 10.1080/14740338.2023.2255524] [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: 07/18/2023] [Accepted: 08/30/2023] [Indexed: 09/07/2023]
Abstract
OBJECTIVES Enzalutamide, a second-generation anti-androgen drug, is an androgen receptor inhibitor developed to overcome resistance to first-generation anti-androgens, such as bicalutamide. This study aimed to identify previously undisclosed adverse events associated with enzalutamide. METHODS Adverse reactions following enzalutamide administration were extracted from the Food and Drug Administration Adverse Event Reporting System (FAERS) database, and the data obtained were from 2014 to 2023. Four algorithms, namely ROR, PRR, BCPNN, and EBGM, were used to detect signs of adverse reactions associated with enzalutamide use. RESULTS This study determined several adverse reactions in the nervous system, including hypogeusia, ageusia, dysgeusia, normal-pressure hydrocephalus, dementia, amnesia, balance disorders, and seizure-like phenomena. The mental aspects manifested as laziness, confusion, and eating disorders. Gastrointestinal system-related adverse reactions included dysphagia, constipation, fecal hardening, and abdominal discomfort. We identified several previously unreported adverse reactions, including normal-pressure hydrocephalus, dementia, balance disorders, eating disorders, and dysphagia. CONCLUSION Our study revealed novel adverse events associated with enzalutamide, particularly in the nervous system, that have not been previously documented. These findings have important implications for future clinical medication guidelines.
Collapse
Affiliation(s)
- Xin Wang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lina Chang
- Shanxi Provincial Hospital of Traditional Chinese Medicine, Department of Physician Standardization Training Center, Taiyuan, Shanxi, China
| | - Zhenhua Li
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuanfang Jiang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yue Chen
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinli Jia
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qiye Wang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaolu Ren
- Shanxi Hospital Affiliated to Cancer Hospital, Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhifang Ma
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wei Zhang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
33
|
Stanojević M, Sollner Dolenc M, Vračko M. Predictive Models for Compound Binding to Androgen and Estrogen Receptors Based on Counter-Propagation Artificial Neural Networks. TOXICS 2023; 11:486. [PMID: 37368586 DOI: 10.3390/toxics11060486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023]
Abstract
Endocrine-disrupting chemicals (EDCs) are exogenous substances that interfere with the normal function of the human endocrine system. These chemicals can affect specific nuclear receptors, such as androgen receptors (ARs) or estrogen receptors (ER) α and β, which play a crucial role in regulating complex physiological processes in humans. It is now more crucial than ever to identify EDCs and reduce exposure to them. For screening and prioritizing chemicals for further experimentation, the use of artificial neural networks (ANN), which allow the modeling of complicated, nonlinear relationships, is most appropriate. We developed six models that predict the binding of a compound to ARs, ERα, or ERβ as agonists or antagonists, using counter-propagation artificial neural networks (CPANN). Models were trained on a dataset of structurally diverse compounds, and activity data were obtained from the CompTox Chemicals Dashboard. Leave-one-out (LOO) tests were performed to validate the models. The results showed that the models had excellent performance with prediction accuracy ranging from 94% to 100%. Therefore, the models can predict the binding affinity of an unknown compound to the selected nuclear receptor based solely on its chemical structure. As such, they represent important alternatives for the safety prioritization of chemicals.
Collapse
Affiliation(s)
- Mark Stanojević
- BiSafe d.o.o., 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | | | - Marjan Vračko
- National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| |
Collapse
|
34
|
Singh VK, Kainat KM, Sharma PK. Crosstalk between epigenetics and tumor promoting androgen signaling in prostate cancer. VITAMINS AND HORMONES 2023; 122:253-282. [PMID: 36863797 DOI: 10.1016/bs.vh.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Prostate cancer (PCa) is one of the major health burdens among all cancer types in men globally. Early diagnosis and efficacious treatment options are highly warranted as far as the incidence of PCa is concerned. Androgen-dependent transcriptional activation of androgen receptor (AR) is central to the prostate tumorigenesis and therefore hormonal ablation therapy remains the first line of treatment for PCa in the clinics. However, the molecular signaling engaged in AR-dependent PCa initiation and progression is infrequent and diverse. Moreover, apart from the genomic changes, non-genomic changes such as epigenetic modifications have also been suggested as critical regulator of PCa development. Among the non-genomic mechanisms, various epigenetic changes such as histones modifications, chromatin methylation and noncoding RNAs regulations etc. play decisive role in the prostate tumorigenesis. Given that epigenetic modifications are reversible using pharmacological modifiers, various promising therapeutic approaches have been designed for the better management of PCa. In this chapter, we discuss the epigenetic control of tumor promoting AR signaling that underlies the mechanism of prostate tumorigenesis and progression. In addition, we have discussed the approaches and opportunities to develop novel epigenetic modifications based therapeutic strategies for targeting PCa including castrate resistant prostate cancer (CRPC).
Collapse
Affiliation(s)
- Vipendra Kumar Singh
- Environmental Carcinogenesis Lab, Food Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India
| | - K M Kainat
- Environmental Carcinogenesis Lab, Food Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Pradeep Kumar Sharma
- Environmental Carcinogenesis Lab, Food Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
35
|
Sheikh IA, Beg MA, Hamoda TAAM, Mandourah HMS, Memili E. Androgen receptor signaling and pyrethroids: Potential male infertility consequences. Front Cell Dev Biol 2023; 11:1173575. [PMID: 37187621 PMCID: PMC10175798 DOI: 10.3389/fcell.2023.1173575] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Infertility is a global health concern inflicting a considerable burden on the global economy and a severe socio-psychological impact. Approximately 15% of couples suffer from infertility globally, with a male factor contribution of approximately 50%. However, male infertility remains largely unexplored, as the burden of infertility is mostly assigned to female people. Endocrine-disrupting chemicals (EDCs) have been proposed as one of the factors causing male infertility. Pyrethroids represent an important class of EDCs, and numerous studies have associated pyrethroid exposure with impaired male reproductive function and development. Therefore, the present study investigated the potentially toxic effects of two common pyrethroids, cypermethrin and deltamethrin, on androgen receptor (AR) signaling. The structural binding characterization of cypermethrin and deltamethrin against the AR ligand-binding pocket was performed using Schrodinger's induced fit docking (IFD) approach. Various parameters were estimated, such as binding interactions, binding energy, docking score, and IFD score. Furthermore, the AR native ligand, testosterone, was subjected to similar experiments against the AR ligand-binding pocket. The results revealed commonality in the amino acid-binding interactions and overlap in other structural parameters between the AR native ligand, testosterone, and the ligands, cypermethrin and deltamethrin. The estimated binding energy values of cypermethrin and deltamethrin were very high and close to those calculated for AR native ligand, testosterone. Taken together, the results of this study suggested potential disruption of AR signaling by cypermethrin and deltamethrin, which may result in androgen dysfunction and subsequent male infertility.
Collapse
Affiliation(s)
- Ishfaq Ahmad Sheikh
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- *Correspondence: Ishfaq Ahmad Sheikh,
| | - Mohd Amin Beg
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | | | - Erdogan Memili
- College of Agriculture and Human Sciences, Prairie View A&M University, Prairie View, TX, United States
| |
Collapse
|
36
|
Suresh PS, Thakur KG, Sharma U. Molecular docking and dynamic simulation approach to decipher steroidal sapogenins (genus Trillium) derived agonists for glucocorticoid receptor. J Biomol Struct Dyn 2023; 41:55-66. [PMID: 34825633 DOI: 10.1080/07391102.2021.2003864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Steroidal sapogenins (SS) are structural analogues of steroidal drugs, which are frequently used for the treatment of several diseases including reproductive, malignancies, neurological, and inflammation-related diseases. The glucocorticoid receptor (GR) is a nuclear receptor that regulates development, metabolism, and inflammation, in response to steroidal ligands. Therefore, GR is considered as a potential therapeutic target for steroidal agents to the treatment of inflammation-related diseases. We hypothesized that SS may act as an agonist for GR due to structural similarity with corticosteroids. In this study, we carried out in silico screening of various SS from the genus Trillium to check their potential as an agonist for GR. Our data suggest that out of 42 SS, only 7 molecules have interacted with GR. However, molecular mechanics with generalized Born and surface area (MM-GBSA) analysis revealed that only two SS (SS 38 and SS 39) molecules bind favorably to GR. Among these, SS 38 (docking score: -9.722 Kcal/mol and MM-GBSA ΔGbind: -50.192 Kcal/mol) and SS 39 (docking score: -11.20 Kcal/mol and MM-GBSA ΔGbind: -58.937 Kcal/mol) have best docking and MM-GBSA scores. Molecular dynamics (MD) simulation studies of SS 38, SS 39, and dexamethasone-GR complex revealed that both SS shows hydrogen bonding and hydrophobic interaction with GR over the 120 ns simulation with mild fluctuations. The current study suggests that SS 38 and SS 39 may be further explored as a potential agonist to treat several disease conditions mediated by GR.
Collapse
Affiliation(s)
- Patil Shivprasad Suresh
- Chemical Technology Division, CSIR-IHBT, Palampur, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P, India
| | - Krishan Gopal Thakur
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P, India.,Structural Biology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Upendra Sharma
- Chemical Technology Division, CSIR-IHBT, Palampur, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P, India
| |
Collapse
|
37
|
Wang Q, Miao J, Zhao A, Wu M, Pan L. Use of GAL4 factor-based yeast assay to quantify the effects of xenobiotics on RXR homodimer and RXR/PPAR heterodimer in scallop Chlamys farreri. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 852:158526. [PMID: 36063929 DOI: 10.1016/j.scitotenv.2022.158526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 06/15/2023]
Abstract
Retinoid X receptor (RXR) and peroxisome proliferators-activated receptors (PPAR) have been shown as important targets of endocrine disrupting effects caused by organotin compounds (OTCs). In vitro methods for non-model species are instrumental in revealing not only mechanism of toxicity but also basic biology. In the present study, we constructed the GAL4 factor-based recombinant yeast systems of RXRα/RXRα (RR), RXRα/PPARα (RPα) and RXRα/PPARγ (RPγ) of the scallop Chlamys farreri to investigate their transcriptional activity under the induction of OTCs (tributyltin chloride, triphenyltin chloride, tripropyltin chloride and bis(tributyltin)oxide), their spiked sediments and five other non‑tin compounds (Wy14643, rosiglitazone, benzyl butyl phthalate, dicyclohexyl phthalate and bis(2-ethylhexyl) phthalate). The results showed that the natural ligand of RXR, 9-cis-retinoic acid (9cRA), induces transcriptional activity in all three systems, while four OTCs induced the transcriptional activity of the RR and RPα systems. None of the five potential non‑tin endocrine disruptors induced effects on the RPα and RPγ systems. The spiked sediment experiment demonstrated the feasibility of the recombinant yeast systems constructed in this study for environmental sample detection. These results suggest that OTCs pose a threat to affect function of RXRα and PPARα of bivalve mollusks. The newly developed GAL4 factor-based yeast two-hybrid system can be used as a valuable tool for identification and quantification of compounds active in disturbing RXR and PPAR of bivalves.
Collapse
Affiliation(s)
- Qiaoqiao Wang
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| | - Jingjing Miao
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China.
| | - Anran Zhao
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| | - Manni Wu
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| | - Luqing Pan
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, PR China
| |
Collapse
|
38
|
Yan L, Rong Q, Zhang H, Jones KC, Li Y, Luo J. Evaluation and Application of a Novel Diffusive Gradients in Thin-Films Technique for In Situ Monitoring of Glucocorticoids in Natural Waters. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:15499-15507. [PMID: 36256587 DOI: 10.1021/acs.est.2c00834] [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: 06/16/2023]
Abstract
The potential environmental risks of glucocorticoids (GCs) have attracted attention in the past few decades. In this study, a novel diffusive gradients in thin-films (DGT) device and analytical technique based on the second generation of polar enhanced phase (PEP-2), PEP-2-DGT, were developed for sampling and quantifying natural and synthetic GCs in aquatic systems. The capacity of PEP-2 gels for accumulating all target compounds was >600 μg per disc, sufficient for long-term passive sampling of selected GCs, even in wastewaters. Systematic tests were carried out to verify the application of DGT in natural waters and wastewaters. The performance of PEP-2-DGT devices was independent (CDGT/Csoln was in the acceptable range of 0.9-1.1) of a wide range of environmental conditions: ionic strength (0.001-0.5 mol L-1), dissolved organic matter (0-20 mg L-1), and pH (3.06-9.02). It was tested for various diffusive layer thicknesses (0.565-2.065 mm) and different deployment times (10-168 h). Diffusion coefficients (D) of selected GCs through an agarose-based diffusive gel were determined for the first time (3.80-4.85 × 10-6 cm-2 s -1 at 25 °C). Linear correlations between D and log Kow were established for three groups of target GCs (R2 = 0.96-0.99). This could enable prediction of D values for other GCs with similar structures in the future, which will help for rapid screening and emergency monitoring. Concentrations and distribution patterns of analytes obtained by PEP-2-DGT devices in five rivers after 7- and 14-day deployments were in accordance with those measured from grab samples, with total GC concentrations ranging from 7 to 27 ng L-1 at all sampling sites, confirming the reliability and robustness of the DGT devices for monitoring GCs in natural waters. The development of the new DGT technique will help improve understanding of the behavior and fate of these compounds in the aquatic environments.
Collapse
Affiliation(s)
- Liying Yan
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu210023, P. R. China
| | - Qiuyu Rong
- Lancaster Environment Centre, Lancaster University, LancasterLA1 4YQ, United Kingdom
| | - Hao Zhang
- Lancaster Environment Centre, Lancaster University, LancasterLA1 4YQ, United Kingdom
| | - Kevin C Jones
- Lancaster Environment Centre, Lancaster University, LancasterLA1 4YQ, United Kingdom
| | - Yanying Li
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu210023, P. R. China
- College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning116023, P. R. China
| | - Jun Luo
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu210023, P. R. China
| |
Collapse
|
39
|
Alemany M. The Roles of Androgens in Humans: Biology, Metabolic Regulation and Health. Int J Mol Sci 2022; 23:11952. [PMID: 36233256 PMCID: PMC9569951 DOI: 10.3390/ijms231911952] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Androgens are an important and diverse group of steroid hormone molecular species. They play varied functional roles, such as the control of metabolic energy fate and partition, the maintenance of skeletal and body protein and integrity and the development of brain capabilities and behavioral setup (including those factors defining maleness). In addition, androgens are the precursors of estrogens, with which they share an extensive control of the reproductive mechanisms (in both sexes). In this review, the types of androgens, their functions and signaling are tabulated and described, including some less-known functions. The close interrelationship between corticosteroids and androgens is also analyzed, centered in the adrenal cortex, together with the main feedback control systems of the hypothalamic-hypophysis-gonads axis, and its modulation by the metabolic environment, sex, age and health. Testosterone (T) is singled out because of its high synthesis rate and turnover, but also because age-related hypogonadism is a key signal for the biologically planned early obsolescence of men, and the delayed onset of a faster rate of functional losses in women after menopause. The close collaboration of T with estradiol (E2) active in the maintenance of body metabolic systems is also presented Their parallel insufficiency has been directly related to the ravages of senescence and the metabolic syndrome constellation of disorders. The clinical use of T to correct hypoandrogenism helps maintain the functionality of core metabolism, limiting excess fat deposition, sarcopenia and cognoscitive frailty (part of these effects are due to the E2 generated from T). The effectiveness of using lipophilic T esters for T replacement treatments is analyzed in depth, and the main problems derived from their application are discussed.
Collapse
Affiliation(s)
- Marià Alemany
- Facultat de Biologia, Universitat de Barcelona, Av. Diagonal, 635, 08028 Barcelona, Catalonia, Spain;
- Institut de Biomedicina, Universitat de Barcelona, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
40
|
Abstract
Embryonic development and cell specification have been viewed as an epigenetically rigid process. Through accumulation of irreversible epigenetic marks, the differentiation process has been considered unidirectional, and once completed cell specification would be permanent and stable. However, somatic cell nuclear transfer that involved the implantation of a somatic nucleus into a previously enucleated oocyte accomplished in amphibians in the 1950s and in mammals in the late 1990s-resulting in the birth of "Dolly the sheep"-clearly showed that "terminal" differentiation is reversible. In parallel, work on lineage-determining factors like MyoD revealed surprising potential to modulate lineage identity in somatic cells. This work culminated in the discovery that a set of four defined factors can reprogram fibroblasts into induced pluripotent stem (iPS) cells, which were shown to be molecularly and functionally equivalent to blastocyst-derived embryonic stem (ES) cells, thus essentially showing that defined factors can induce authentic reprogramming without the need of oocytes. This concept was further extended when it was shown that fibroblasts can be directly converted into neurons, showing induced lineage conversion is possible even between cells representing two different germ layers. These findings suggest that "everything is possible" (i.e., once key lineage reprogramming factors are identified, cells should be able to convert into any desired lineage).
Collapse
Affiliation(s)
- Hannah Shelby
- Departments of Pathology and Chemical and Systems Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Tara Shelby
- Departments of Pathology and Chemical and Systems Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Marius Wernig
- Departments of Pathology and Chemical and Systems Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
41
|
Johnson RP, Ratnacaram CK, Kumar L, Jose J. Combinatorial approaches of nanotherapeutics for inflammatory pathway targeted therapy of prostate cancer. Drug Resist Updat 2022; 64:100865. [PMID: 36099796 DOI: 10.1016/j.drup.2022.100865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PC) is the most prevalent male urogenital cancer worldwide. PC patients presenting an advanced or metastatic cancer succumb to the disease, even after therapeutic interventions including radiotherapy, surgery, androgen deprivation therapy (ADT), and chemotherapy. One of the hallmarks of PC is evading immune surveillance and chronic inflammation, which is a major challenge towards designing effective therapeutic formulations against PC. Chronic inflammation in PC is often characterized by tumor microenvironment alterations, epithelial-mesenchymal transition and extracellular matrix modifications. The inflammatory events are modulated by reactive nitrogen and oxygen species, inflammatory cytokines and chemokines. Major signaling pathways in PC includes androgen receptor, PI3K and NF-κB pathways and targeting these inter-linked pathways poses a major therapeutic challenge. Notably, many conventional treatments are clinically unsuccessful, due to lack of targetability and poor bioavailability of the therapeutics, untoward toxicity and multidrug resistance. The past decade witnessed an advancement of nanotechnology as an excellent therapeutic paradigm for PC therapy. Modern nanovectorization strategies such as stimuli-responsive and active PC targeting carriers offer controlled release patterns and superior anti-cancer effects. The current review initially describes the classification, inflammatory triggers and major inflammatory pathways of PC, various PC treatment strategies and their limitations. Subsequently, recent advancement in combinatorial nanotherapeutic approaches, which target PC inflammatory pathways, and the mechanism of action are discussed. Besides, the current clinical status and prospects of PC homing nanovectorization, and major challenges to be addressed towards the advancement PC therapy are also addressed.
Collapse
Affiliation(s)
- Renjith P Johnson
- Polymer Nanobiomaterial Research Laboratory, Nanoscience and Microfluidics Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Chandrahas Koumar Ratnacaram
- Cell Signaling and Cancer Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Lalit Kumar
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka 576 104, India
| | - Jobin Jose
- NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangalore 575018, India.
| |
Collapse
|
42
|
Avgeris I, Pliatsika D, Nikolaropoulos SS, Fousteris MA. Targeting androgen receptor for prostate cancer therapy: From small molecules to PROTACs. Bioorg Chem 2022; 128:106089. [PMID: 35973305 DOI: 10.1016/j.bioorg.2022.106089] [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] [Received: 05/09/2022] [Revised: 07/30/2022] [Accepted: 08/06/2022] [Indexed: 12/13/2022]
Abstract
Prostate cancer (PCa) remains a serious type of cancer for men worldwide. The majority of new PCa cases are associated with androgen receptor (AR) hyperactivity. Various AR-targeting molecules that suppress its activity have been discovered. In this review, we present the already marketed antiandrogens and a selection of structurally and chemically interesting AR-targeting compounds, from a pharmacochemical perspective. Focus has been placed on the applied design approaches, structural evolution and structure-activity relationships of the most prominent compound classes. Passing from the traditional steroidal AR antagonists to the modern AR-targeting proteolysis targeting chimeras (PROTACs), we intend to provide a comprehensive overview on AR-targeting molecules for PCa treatment.
Collapse
Affiliation(s)
- Ioannis Avgeris
- Laboratory of Medicinal Chemistry, Department of Pharmacy, University of Patras, Patras GR-26500, Greece
| | - Dimanthi Pliatsika
- Laboratory of Medicinal Chemistry, Department of Pharmacy, University of Patras, Patras GR-26500, Greece
| | - Sotiris S Nikolaropoulos
- Laboratory of Medicinal Chemistry, Department of Pharmacy, University of Patras, Patras GR-26500, Greece
| | - Manolis A Fousteris
- Laboratory of Medicinal Chemistry, Department of Pharmacy, University of Patras, Patras GR-26500, Greece.
| |
Collapse
|
43
|
TMAO to the rescue of pathogenic protein variants. Biochim Biophys Acta Gen Subj 2022; 1866:130214. [PMID: 35902028 DOI: 10.1016/j.bbagen.2022.130214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/11/2022] [Accepted: 07/21/2022] [Indexed: 11/22/2022]
Abstract
Trimethylamine N-oxide (TMAO) is a chemical chaperone found in various organisms including humans. Various studies unveiled that it is an excellent protein-stabilizing agent, and induces folding of unstructured proteins. It is also well established that it can counteract the deleterious effects of urea, salt, and hydrostatic pressure on macromolecular integrity. There is also existence of large body of data regarding its ability to restore functional deficiency of various mutant proteins or pathogenic variants by correcting misfolding defects and inhibiting the formation of high-order toxic protein oligomers. Since an important class of human disease called "protein conformational disorders" is due to protein misfolding and/or formation of high-order oligomers, TMAO stands as a promising molecule for the therapeutic intervention of such diseases. The present review has been designed to gather a comprehensive knowledge of the TMAO's effect on the functional restoration of various mutants, identify its shortcomings and explore its potentiality as a lead molecule. Future prospects have also been suitably incorporated.
Collapse
|
44
|
Zhang L, Zhang J, Nie Q. DIRECT-NET: An efficient method to discover cis-regulatory elements and construct regulatory networks from single-cell multiomics data. SCIENCE ADVANCES 2022; 8:eabl7393. [PMID: 35648859 PMCID: PMC9159696 DOI: 10.1126/sciadv.abl7393] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 04/14/2022] [Indexed: 05/13/2023]
Abstract
The emergence of single-cell multiomics data provides unprecedented opportunities to scrutinize the transcriptional regulatory mechanisms controlling cell identity. However, how to use those datasets to dissect the cis-regulatory element (CRE)-to-gene relationships at a single-cell level remains a major challenge. Here, we present DIRECT-NET, a machine-learning method based on gradient boosting, to identify genome-wide CREs and their relationship to target genes, either from parallel single-cell gene expression and chromatin accessibility data or from single-cell chromatin accessibility data alone. By extensively evaluating and characterizing DIRECT-NET's predicted CREs using independent functional genomics data, we find that DIRECT-NET substantially improves the accuracy of inferring CRE-to-gene relationships in comparison to existing methods. DIRECT-NET is also capable of revealing cell subpopulation-specific and dynamic regulatory linkages. Overall, DIRECT-NET provides an efficient tool for predicting transcriptional regulation codes from single-cell multiomics data.
Collapse
Affiliation(s)
- Lihua Zhang
- School of Computer Science, Wuhan University, Wuhan 430072, China
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Jing Zhang
- Department of Computer Science, University of California, Irvine, Irvine, CA 92697, USA
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
45
|
Das K, Patil A, Goren A, Cockerell CJ, Goldust M. Androgens and COVID-19. J Cosmet Dermatol 2022; 21:3176-3180. [PMID: 35576054 PMCID: PMC9348029 DOI: 10.1111/jocd.15090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022]
Abstract
Background The humans have been disproportionately affected by the coronavirus disease (COVID‐19) pandemic. The novel coronavirus or the severe acute respiratory syndrome coronavirus 2 (SARS‐COV2) causing coronavirus disease (COVID‐19) has spread across the globe. Androgens have been suggested to have a role in COVID‐19 pathogenesis. Objective The objective of this review article is to study the link between androgens and COVID‐19. Methodology PubMed and Google Scholar search was performed to retrieve literature related to the topic. Review articles, clinical trials, retrospective studies, observational studies, and case–control studies were considered for the review. Results Severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) infected men are more inclined to be hospitalized for intensive care unit (ICU) as compared with women. This difference in the ICU admissions provides some clue for possible influence of androgens in the severity of COVID‐19. The contribution of androgen and androgen receptor in COVID‐19 disease and its severity, as well as the numerous medications targeting androgen and its receptor for lowering COVID‐19 disease severity, are discussed in this review. Available literature suggests the role of androgen in the pathogenesis and severity of COVID‐19. Sensitivity for androgen may be an important factor in regulating the severity of COVID‐19 disease. Conclusion There is a scope for the development of COVID‐19 treatments based on androgen suppression. Clinical trials may furnish pivotal data and add more evidence‐based options for the management of COVID‐19.
Collapse
Affiliation(s)
- Kinnor Das
- Consultant Dermatolgist, Apollo clinic, Silchar, Assam, India
| | - Anant Patil
- Department of Pharmacology, Dr. DY Patil Medical College, Navi Mumbai, India
| | - Andy Goren
- University of Rome G. Marconi, Rome, Italy
| | - Clay J Cockerell
- Departments of Dermatology and Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Cockerell Dermatopathology, Dallas, Texas, USA
| | - Mohamad Goldust
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
46
|
LaFever BJ, Kawasawa YI, Ito A, Imamura F. Pathological consequences of chronic olfactory inflammation on neurite morphology of olfactory bulb projection neurons. Brain Behav Immun Health 2022; 21:100451. [PMID: 35360408 PMCID: PMC8960895 DOI: 10.1016/j.bbih.2022.100451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 11/22/2022] Open
Abstract
Chronic olfactory inflammation (COI) in conditions such as chronic rhinosinusitis significantly impairs the functional and anatomical components of the olfactory system. COI induced by intranasal administration of lipopolysaccharide (LPS) results in atrophy, gliosis, and pro-inflammatory cytokine production in the olfactory bulb (OB). Although chronic rhinosinusitis patients have smaller OBs, the consequences of olfactory inflammation on OB neurons are largely unknown. In this study, we investigated the neurological consequences of COI on OB projection neurons, mitral cells (MCs) and tufted cells (TCs). To induce COI, we performed unilateral intranasal administration of LPS to mice for 4 and 10 weeks. Effects of COI on the OB were examined using RNA-sequencing approaches and immunohistochemical analyses. We found that repeated LPS administration upregulated immune-related biological pathways in the OB after 4 weeks. We also determined that the length of TC lateral dendrites in the OB significantly decreased after 10 weeks of COI. The axon initial segment of TCs decreased in number and in length after 10 weeks of COI. The lateral dendrites and axon initial segments of MCs, however, were largely unaffected. In addition, dendritic arborization and AIS reconstruction both took place following a 10-week recovery period. Our findings suggest that olfactory inflammation specifically affects TCs and their integrated circuitry, whereas MCs are potentially protected from this condition. This data demonstrates unique characteristics of the OBs ability to undergo neuroplastic changes in response to stress.
Early-stage chronic olfactory inflammation activates the interferon-γ-driven inflammatory pathways in the olfactory bulb. Tufted cells undergo neurite dysregulation in response to chronic olfactory inflammation. Mitral cells and interneurons in the external plexiform layer are largely unaffected by chronic olfactory inflammation. Tufted cells experience complete recovery from neurite dysregulation following a period of ceased inflammation
Collapse
Affiliation(s)
- Brandon J. LaFever
- Department of Pharmacology, Penn State College of Medicine, 500 University Dr., Hershey, PA, 17033, USA
| | - Yuka Imamura Kawasawa
- Department of Pharmacology, Penn State College of Medicine, 500 University Dr., Hershey, PA, 17033, USA
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, 500 University Dr., Hershey, PA, 17033, USA
- Institute for Personalized Medicine, Penn State College of Medicine, 500 University Dr., Hershey, PA, 17033, USA
| | - Ayako Ito
- Department of Pharmacology, Penn State College of Medicine, 500 University Dr., Hershey, PA, 17033, USA
| | - Fumiaki Imamura
- Department of Pharmacology, Penn State College of Medicine, 500 University Dr., Hershey, PA, 17033, USA
- Corresponding author. Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA, 17033, USA.
| |
Collapse
|
47
|
Sharp JL, Pearson T, Smith MA. Sex differences in opioid receptor mediated effects: Role of androgens. Neurosci Biobehav Rev 2022; 134:104522. [PMID: 34995646 PMCID: PMC8872632 DOI: 10.1016/j.neubiorev.2022.104522] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/19/2021] [Accepted: 01/02/2022] [Indexed: 12/26/2022]
Abstract
An abundance of data indicates there are sex differences in endogenous opioid peptides and opioid receptors, leading to functional differences in sensitivity to opioid receptor mediated behaviors between males and females. Many of these sex differences are mediated by the effects of gonadal hormones on the endogenous opioid system. Whereas much research has examined the role of ovarian hormones on opioid receptor mediated endpoints, comparatively less research has examined the role of androgens. This review describes what is currently known regarding the influence of androgens on opioid receptor mediated endpoints and how androgens may contribute to sex differences in these effects. The review also addresses the clinical implications of androgenic modulation of opioid receptor mediated behaviors and suggests future lines of research for preclinical and clinical investigators. We conclude that further investigation into androgenic modulation of opioid receptor mediated effects may lead to new options for addressing conditions such as chronic pain and substance use disorders.
Collapse
Affiliation(s)
- Jessica L Sharp
- Department of Psychology and Program in Neuroscience, Davidson College, United States
| | - Tallia Pearson
- Department of Psychology and Program in Neuroscience, Davidson College, United States
| | - Mark A Smith
- Department of Psychology and Program in Neuroscience, Davidson College, United States.
| |
Collapse
|
48
|
Marcheva B, Weidemann BJ, Taguchi A, Perelis M, Ramsey KM, Newman MV, Kobayashi Y, Omura C, Manning Fox JE, Lin H, Macdonald PE, Bass J. P2Y1 purinergic receptor identified as a diabetes target in a small-molecule screen to reverse circadian β-cell failure. eLife 2022; 11:e75132. [PMID: 35188462 PMCID: PMC8860442 DOI: 10.7554/elife.75132] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/21/2022] [Indexed: 12/18/2022] Open
Abstract
The mammalian circadian clock drives daily oscillations in physiology and behavior through an autoregulatory transcription feedback loop present in central and peripheral cells. Ablation of the core clock within the endocrine pancreas of adult animals impairs the transcription and splicing of genes involved in hormone exocytosis and causes hypoinsulinemic diabetes. Here, we developed a genetically sensitized small-molecule screen to identify druggable proteins and mechanistic pathways involved in circadian β-cell failure. Our approach was to generate β-cells expressing a nanoluciferase reporter within the proinsulin polypeptide to screen 2640 pharmacologically active compounds and identify insulinotropic molecules that bypass the secretory defect in CRISPR-Cas9-targeted clock mutant β-cells. We validated hit compounds in primary mouse islets and identified known modulators of ligand-gated ion channels and G-protein-coupled receptors, including the antihelmintic ivermectin. Single-cell electrophysiology in circadian mutant mouse and human cadaveric islets revealed ivermectin as a glucose-dependent secretagogue. Genetic, genomic, and pharmacological analyses established the P2Y1 receptor as a clock-controlled mediator of the insulinotropic activity of ivermectin. These findings identify the P2Y1 purinergic receptor as a diabetes target based upon a genetically sensitized phenotypic screen.
Collapse
Affiliation(s)
- Biliana Marcheva
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Benjamin J Weidemann
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Akihiko Taguchi
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of MedicineChicagoUnited States
- Division of Endocrinology, Metabolism, Hematological Science and Therapeutics, Department of Bio-Signal Analysis, Yamaguchi University, Graduate School of Medicine, 1-1-1YamaguchiJapan
| | - Mark Perelis
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of MedicineChicagoUnited States
- Ionis Pharmaceuticals, IncCarlsbadUnited States
| | - Kathryn Moynihan Ramsey
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Marsha V Newman
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Yumiko Kobayashi
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Chiaki Omura
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Jocelyn E Manning Fox
- Department of Pharmacology, Alberta Diabetes Institute, University of AlbertaEdmonton, ABCanada
| | - Haopeng Lin
- Department of Pharmacology, Alberta Diabetes Institute, University of AlbertaEdmonton, ABCanada
| | - Patrick E Macdonald
- Department of Pharmacology, Alberta Diabetes Institute, University of AlbertaEdmonton, ABCanada
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of MedicineChicagoUnited States
| |
Collapse
|
49
|
Chen MK, Liang ZJ, Luo DS, Xue KY, Liao DY, Li Z, Yu Y, Chen ZS, Zhao SC. Abiraterone, Orteronel, Enzalutamide and Docetaxel: Sequential or Combined Therapy? Front Pharmacol 2022; 13:843110. [PMID: 35250590 PMCID: PMC8891580 DOI: 10.3389/fphar.2022.843110] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
Objective: To summarize the current therapeutic status using chemotherapeutic agent docetaxel and endocrine therapeutic agents (ARAT, abiraterone, orteronel or enzalutamide) for the treatment of metastatic castration-resistant prostate cancer (mCRPC), including sequential therapy and combined therapy, to promote the consensus on the optimal regimen for achieving superior treatment efficacy.Methods: Through literature search in PubMed, articles with the following relevant keywords were collected and anlyzed: CRPC, abiraterone, orteronel and enzalutamide, median survival, overall survival, prostate specific antigen (PSA), PSA response rate and median radiologic progression-free survival.Results: Fifty-eight articles were obtained and analyzed in this review. These articles included androgen axis-targeting agents after docetaxel, docetaxel after androgen axis-targeting agents, Triple sequential and combination therapy, covering four current drugs for mCRPC treatment: docetaxel, abiraterone, orteronel, and enzalutamide. It was found that there may be some cross-resistance between androgen axis-targeting agents, which will reduce the efficacy of subsequent drug treatment. Although neither of the studies of using combination therapy showed serious drug toxicity, the efficacy of sequential therapy was not as good as expected. Most adverse reactions after treatment were reported to be level 1–2.Conclusion: Based on the results of the current studies, abiraterone followed by enzalutamide treatment is the best sequential treatment for most docetaxel-naïve patients. This treatment achieves not only good OS, but also PFS and PSA response rates. In addition, for patients who have previously failed docetaxel treatment, enzalutamide is the best choice as the subsequent treatment.
Collapse
Affiliation(s)
- Ming-kun Chen
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Urology, The Third Clinical College of Southern Medical University, Guangzhou, China
| | - Zhi-jian Liang
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Urology, The Third Clinical College of Southern Medical University, Guangzhou, China
| | - Dao-Sheng Luo
- Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Kang-yi Xue
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Urology, The Third Clinical College of Southern Medical University, Guangzhou, China
| | - De-ying Liao
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Urology, The Third Clinical College of Southern Medical University, Guangzhou, China
| | - Zheshen Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Yuzhong Yu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
- *Correspondence: Zhe-Sheng Chen, ; Shan-Chao Zhao,
| | - Shan-Chao Zhao
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Department of Urology, The Third Clinical College of Southern Medical University, Guangzhou, China
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Zhe-Sheng Chen, ; Shan-Chao Zhao,
| |
Collapse
|
50
|
Bondos SE, Dunker AK, Uversky VN. Intrinsically disordered proteins play diverse roles in cell signaling. Cell Commun Signal 2022; 20:20. [PMID: 35177069 PMCID: PMC8851865 DOI: 10.1186/s12964-022-00821-7] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/11/2021] [Indexed: 11/29/2022] Open
Abstract
Signaling pathways allow cells to detect and respond to a wide variety of chemical (e.g. Ca2+ or chemokine proteins) and physical stimuli (e.g., sheer stress, light). Together, these pathways form an extensive communication network that regulates basic cell activities and coordinates the function of multiple cells or tissues. The process of cell signaling imposes many demands on the proteins that comprise these pathways, including the abilities to form active and inactive states, and to engage in multiple protein interactions. Furthermore, successful signaling often requires amplifying the signal, regulating or tuning the response to the signal, combining information sourced from multiple pathways, all while ensuring fidelity of the process. This sensitivity, adaptability, and tunability are possible, in part, due to the inclusion of intrinsically disordered regions in many proteins involved in cell signaling. The goal of this collection is to highlight the many roles of intrinsic disorder in cell signaling. Following an overview of resources that can be used to study intrinsically disordered proteins, this review highlights the critical role of intrinsically disordered proteins for signaling in widely diverse organisms (animals, plants, bacteria, fungi), in every category of cell signaling pathway (autocrine, juxtacrine, intracrine, paracrine, and endocrine) and at each stage (ligand, receptor, transducer, effector, terminator) in the cell signaling process. Thus, a cell signaling pathway cannot be fully described without understanding how intrinsically disordered protein regions contribute to its function. The ubiquitous presence of intrinsic disorder in different stages of diverse cell signaling pathways suggest that more mechanisms by which disorder modulates intra- and inter-cell signals remain to be discovered.
Collapse
Affiliation(s)
- Sarah E. Bondos
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843 USA
| | - A. Keith Dunker
- Center for Computational Biology and Bioinformatics, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Vladimir N. Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612 USA
- Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Pushchino, Moscow Region, Russia 142290
| |
Collapse
|