1
|
Liu Y, Ning Y, Jiang Y, Ou Y, Chen X, Zhong C, Wang R, Zhang Z, Wang K, Long D, Zhao W. Effects of combined exposure to polyethylene and oxidized polycyclic aromatic hydrocarbons on growth, development, and neurobehavior in Zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 283:107361. [PMID: 40233682 DOI: 10.1016/j.aquatox.2025.107361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/05/2025] [Accepted: 04/09/2025] [Indexed: 04/17/2025]
Abstract
Oxygenated polycyclic aromatic hydrocarbons (OPAHs) are a class of anthropogenic, persistent, and highly toxic PAH contaminants associated with developmental toxicity, 9-fluorenone (9-FLO) is a typical member of the OPAH family. Due to its ketone group, it has higher polarity, which results in increased solubility in water and greater potential for transport via atmospheric particles or water bodies. Polyethylene (PE), an amorphous polymer, is characterized by high diffusivity, high permeability, and a large internal molecular free volume, which confers a strong absorption capacity for organic pollutants. The effects of individual and combined exposures to these two common environmental pollutants on aquatic life remain unclear. In this study, we evaluated the effects of PE and 9-FLO exposure on growth, development, metabolism, and behavior using zebrafish as a model organism. We employed methods and techniques such as acridine orange staining, enzyme-linked immunosorbent assay (ELISA), video tracking, automated behavior analysis, microscopy imaging, and real-time fluorescence quantification. Zebrafish embryos at 2 h post-fertilization (hpf) were exposed to PE and 9-FLO, both individually and in combination. Our studies showed that exposure to PE or 9-FLO alone increases embryonic mortality and decreases hatchability compared to the control group. The 9-FLO group exhibited delayed hatching and inhibited larval length growth. The exposed groups showed a loose arrangement of telencephalic neurons, partial apoptosis, decreased dopamine (DA) content, increased serotonin (5-HT) content, decreased exercise capacity, reduced rhythmic amplitude, and increased rest time. The combined exposure group showed a slight alleviation of these effects compared to the single exposure groups but still exhibited significant differences from the control group. In summary, early exposure to PE and 9-FLO in zebrafish embryos, whether alone or in combination, affects growth, development, apoptosis, neurotransmitter release, and motor behavior of zebrafish neurons.
Collapse
Affiliation(s)
- Yu Liu
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yujun Ning
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yi Jiang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Yiquan Ou
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China; Department of nutrition, The First People's Hospital of Chenzhou, Chenzhou No.1 People's Hospital, Chenzhou 423000, China
| | - Xiaobing Chen
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Chiting Zhong
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Ru Wang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Zhibo Zhang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China; First Clinical Faculty, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Kongfan Wang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Dingxin Long
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Weichao Zhao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
2
|
Bakr S, Dash HE, Youssef AAA, El-Hamid RGA. Differential Expression of BCL2 and IGFBP2 in Childhood Immune Thrombocytopenic Purpura Clinical Subtypes: Implications for Predicting Disease Progression and Apoptotic Regulation. Pediatr Blood Cancer 2025; 72:e31586. [PMID: 39905592 DOI: 10.1002/pbc.31586] [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: 11/11/2024] [Revised: 01/03/2025] [Accepted: 01/28/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Immune thrombocytopenic purpura (ITP), which poses challenges in treatment response, is an autoimmune-mediated bleeding disorder with an extremely complex pathogenesis and unpredictable clinical progression. Dysregulation of apoptotic pathways may influence both the pathogenesis and prognosis of ITP. This study aimed to evaluate the expression patterns of the apoptotic protein insulin-like growth factor-binding protein 2 (IGFBP2) and the anti-apoptotic protein B-cell lymphoma 2 (BCL2) as potential predictive or prognostic biomarkers for disease progression in childhood ITP. PATIENTS AND METHODS The expression levels of BCL2 and IGFBP2 were assessed in peripheral blood samples from 40 pediatric ITP patients and 30 age- and sex-matched healthy controls using enzyme-linked immunosorbent assays. RESULTS Plasma levels of BCL2 and IGFBP2 were higher in ITP patients than in control subjects. Although the difference in IGFBP2 expression was not statistically significant (p = 0.910), BCL2 expression was significantly elevated (p < 0.001). Notably, chronic ITP patients had significantly lower levels of both IGFBP2 and BCL2 markers compared to patients who achieved spontaneous recovery (p < 0.001). CONCLUSION BCL2 and IGFBP2 appear to be promising noninvasive biomarkers for predicting disease outcomes in newly diagnosed ITP, emphasizing the need for validation in large-scale, multicenter longitudinal studies.
Collapse
Affiliation(s)
- Salwa Bakr
- Department of Clinical Pathology/Hematology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Hanaa El Dash
- Department of Pediatric, Fayoum University, Fayoum, Egypt
| | | | | |
Collapse
|
3
|
He W, Loganathan N, Belsham DD. IGF1 Signaling Regulates Neuropeptide Expression in Hypothalamic Neurons Under Physiological and Pathological Conditions. Endocrinology 2025; 166:bqaf051. [PMID: 40105689 PMCID: PMC11949690 DOI: 10.1210/endocr/bqaf051] [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: 01/29/2025] [Revised: 02/28/2025] [Accepted: 03/17/2025] [Indexed: 03/20/2025]
Abstract
Insulin-like growth factor 1 (IGF1) plays a critical role in metabolism and aging, but its role in the brain remains unclear. This study examined whether hypothalamic neurons respond to IGF1 and how its actions are modulated. RT-qPCR and single-cell RNA sequencing indicated that Igf1r mRNA is expressed in neuropeptide Y/Agouti-related peptide (NPY/AgRP) neurons but has higher expression in pro-opiomelanocortin (POMC) neurons. IGF1 binding proteins Igfbp3 and Igfbp5 were significantly expressed, whereby Igfbp5 levels were modulated by fasting, nutrient availability, and circadian rhythms, implying that IGF1 signaling can be controlled by multiple mechanisms. In mouse and human models, IGF1 regulated Agrp, Npy, Pomc, Cartpt, Spx, Gal, and Fam237b expression, producing an overall anorexigenic profile. Hyperinsulinemia induced IGF1 resistance, accompanied by reduced IGF1R protein, as well as Igf1r and Irs2 mRNA expression via over-activation of phosphoinositide 3-kinase/forkhead box O1 (PI3K-FOXO1) signaling. Thus, hypothalamic neurons respond to IGF1 under physiological conditions, and hyperinsulinemia is a novel mechanism that drives cellular IGF1 resistance.
Collapse
Affiliation(s)
- Wenyuan He
- Department of Physiology, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Neruja Loganathan
- Department of Physiology, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Denise D Belsham
- Department of Physiology, University of Toronto, Toronto, ON, Canada M5S 1A8
- Department of Medicine, University of Toronto, Toronto, ON, Canada M5S 1A8
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada M5S 1A8
| |
Collapse
|
4
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [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: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
5
|
Fernández-Pereira C, Agís-Balboa RC. The Insulin-like Growth Factor Family as a Potential Peripheral Biomarker in Psychiatric Disorders: A Systematic Review. Int J Mol Sci 2025; 26:2561. [PMID: 40141202 PMCID: PMC11942524 DOI: 10.3390/ijms26062561] [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: 12/30/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 03/28/2025] Open
Abstract
Psychiatric disorders (PDs), including schizophrenia (SZ), major depressive disorder (MDD), bipolar disorder (BD), autism spectrum disorder (ASD), among other disorders, represent a significant global health burden. Despite advancements in understanding their biological mechanisms, there is still no reliable objective and reliable biomarker; therefore, diagnosis remains largely reliant on subjective clinical assessments. Peripheral biomarkers in plasma or serum are interesting due to their accessibility, low cost, and potential to reflect central nervous system processes. Among these, the insulin-like growth factor (IGF) family, IGF-1, IGF-2, and IGF-binding proteins (IGFBPs), has gained attention for its roles in neuroplasticity, cognition, and neuroprotection, as well as for their capability to cross the blood-brain barrier. This review evaluates the evidence for IGF family alterations in PDs, with special focus on SZ, MDD, and BD, while also addressing other PDs covering almost 40 years of history. In SZ patients, IGF-1 alterations have been linked to metabolic dysregulation, treatment response, and hypothalamic-pituitary-adrenal axis dysfunction. In MDD patients, IGF-1 appears to compensate for impaired neurogenesis, although findings are inconsistent. Emerging studies on IGF-2 and IGFBPs suggest potential roles across PDs. While promising, heterogeneity among studies and methodological limitations highlights the need for further research to validate IGFs as reliable psychiatric biomarkers.
Collapse
Affiliation(s)
- Carlos Fernández-Pereira
- Neuro Epigenetics Lab, Health Research Institute of Santiago de Compostela (IDIS), Santiago University Hospital Complex, 15706 Santiago de Compostela, Spain;
- Translational Research in Neurological Diseases (ITEN) Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago University Hospital Complex, SERGAS-USC, 15706 Santiago de Compostela, Spain
- Neurology Service, Santiago University Hospital Complex, 15706 Santiago de Compostela, Spain
| | - Roberto Carlos Agís-Balboa
- Neuro Epigenetics Lab, Health Research Institute of Santiago de Compostela (IDIS), Santiago University Hospital Complex, 15706 Santiago de Compostela, Spain;
- Translational Research in Neurological Diseases (ITEN) Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago University Hospital Complex, SERGAS-USC, 15706 Santiago de Compostela, Spain
- Neurology Service, Santiago University Hospital Complex, 15706 Santiago de Compostela, Spain
| |
Collapse
|
6
|
Shi L, Ding Z, Chen J. Deciphering the role of IGFBP5 in delaying fibrosis and sarcopenia in aging skeletal muscle: therapeutic implications and molecular mechanisms. Front Pharmacol 2025; 16:1557703. [PMID: 40144669 PMCID: PMC11937025 DOI: 10.3389/fphar.2025.1557703] [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: 01/09/2025] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
Introduction Sarcopenia is a condition characterized by the loss of muscle fibers and excessive deposition of extracellular matrix proteins. The interplay between muscle atrophy and fibrosis is a central feature of sarcopenia. While the mechanisms underlying skeletal muscle aging and fibrosis remain incompletely understood, cellular senescence has emerged as a key contributor. This study investigates the role of D-galactose (D-gal) in inducing fibroblasts senescence and skeletal muscle fibrosis, and aims to find the key regulator of the process to serve as a therapeutical target. Methods To discover the role of D-gal in inducing cellular senescence and fibrosis, the senescence markers and the expression of fibrosis-related proteins were assessed after introducing D-gal among fibroblasts, and muscle strength and mass. The severity of muscle atrophy and fibrosis were also verified by using H&E staining and Masson trichrome staining after D-gal treatment via subcutaneous injection among mice. Subsequently, mRNA sequencing (RNA-seq) was performed and the differential expressed genes were identified between under D-gal or control treatment, to discover the key regulator of D-GAL-driven fibroblasts senescence and fibrosis. The role of the key regulator IGFBP5 were then validated in D-GAL treated IGFBP5-knockdown fibroblasts in vitro by analyzing the level of senescence and fibrosis-related markers. And the results were further confirmed in vivo in IGFBP5-knockdown SAMP8 mice with histological examinations. Results D-gal treatment effectively induced cellular senescence and fibrosis in fibroblasts, as well as skeletal muscle atrophy, fibrosis and loss in muscle mass and function in mice. IGFBP5 was identified as a key regulator of D-GAL induced senescence and fibrosis among fibroblasts using RNA-seq. And further validation tests showed that IGFBP5-knockdown could alleviate D-GAL-induced fibroblast cellular senescence and fibrosis, as well as the severity of muscle atrophy and fibrosis in SAMP8 mice. Discussion IGFBP5 emerging as a key regulator of D-GAL-induced fibroblast cellular senescence and fibrosis. The findings provide new insights into the molecular mechanisms underlying age-related skeletal muscle fibrosis and highlight IGFBP5 as a potential therapeutic target. Further research is needed to validate these findings and explore related clinical applications.
Collapse
Affiliation(s)
| | - Zheci Ding
- *Correspondence: Zheci Ding, ; Jiwu Chen,
| | - Jiwu Chen
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
7
|
Conover CA, Oxvig C. The IGF System and Aging. Endocr Rev 2025; 46:214-223. [PMID: 39418083 PMCID: PMC11894535 DOI: 10.1210/endrev/bnae029] [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: 07/03/2024] [Revised: 09/20/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
There is strong evidence that IGF signaling is involved in fundamental aspects of the aging process. However, the extracellular part of the IGF system is complex with various receptors, ligand effectors, high-affinity IGF-binding proteins, proteinases, and endogenous inhibitors that all, along with their biological context, must be considered. The IGF system components are evolutionarily conserved, underscoring the importance of understanding this system in physiology and pathophysiology. This review will briefly describe the different components of the IGF system and then discuss past and current literature regarding IGF and aging, with a focus on cellular senescence, model organisms of aging, centenarian genetics, and 3 age-related diseases-pulmonary fibrosis, Alzheimer disease, and macular degeneration-in appropriate murine models and in humans. Commonalities in mechanism suggest conditions where IGF system components may be disease drivers and potential targets in promoting healthy aging in humans.
Collapse
Affiliation(s)
- Cheryl A Conover
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
8
|
Dev W, Sultana F, Li H, Hu D, Peng Z, He S, Zhang H, Waqas M, Geng X, Du X. Molecular mechanisms of cold stress response in cotton: Transcriptional reprogramming and genetic strategies for tolerance. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2025; 352:112390. [PMID: 39827949 DOI: 10.1016/j.plantsci.2025.112390] [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: 09/27/2024] [Revised: 01/07/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Cold stress has a huge impact on the growth and development of cotton, presenting a significant challenge to its productivity. Comprehending the complex molecular mechanisms that control the reaction to CS is necessary for developing tactics to improve cold tolerance in cotton. This review paper explores how cotton responds to cold stress by regulating gene expression, focusing on both activating and repressing specific genes. We investigate the essential roles that transcription factors and regulatory elements have in responding to cold stress and controlling gene expression to counteract the negative impacts of low temperatures. Through a comprehensive examination of new publications, we clarify the intricacies of transcriptional reprogramming induced by cold stress, emphasizing the connections between different regulatory elements and signaling pathways. Additionally, we investigate the consecutive effects of cold stress on cotton yield, highlighting the physiological and developmental disturbances resulting from extended periods of low temperatures. The knowledge obtained from this assessment allows for a more profound comprehension of the molecular mechanisms that regulate cold stress responses, suggesting potential paths for future research to enhance cold tolerance in cotton by utilizing targeted genetic modifications and biotechnological interventions.
Collapse
Affiliation(s)
- Washu Dev
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Fahmida Sultana
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Hongge Li
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China; National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya, Hainan 57202, China
| | - Daowu Hu
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China; National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya, Hainan 57202, China
| | - Zhen Peng
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Shoupu He
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Haobo Zhang
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Muhammad Waqas
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Xiaoli Geng
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Xiongming Du
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China; National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya, Hainan 57202, China.
| |
Collapse
|
9
|
Tam V, Chopra N, Sima S, Chen P, Sharma R, Chan D, Diwan A. Effects of GDF6 on active protein synthesis by cells of degenerated intervertebral disc. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2025:10.1007/s00586-025-08715-1. [PMID: 39920317 DOI: 10.1007/s00586-025-08715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/09/2025]
Abstract
INTRODUCTION Intervertebral disc degeneration (IVD) is a leading cause of low back pain, a prevalent musculoskeletal condition. IVD degeneration is characterized by the degradation of nucleus pulposus (NP), annulus fibrosus (AF), and cartilage endplates (EP). Growth Differentiation Factor 6 (GDF6), part of the bone morphogenetic protein family, has demonstrated potential in maintaining disc integrity. However, its precise role in cellular protein synthesis during IVD degeneration remains unclear. METHODS This study employed Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) to investigate the effects of GDF6 on protein synthesis in NP, AF, and EP cells isolated from degenerated human IVDs. Cells were cultured in SILAC media with and without GDF6 treatment. The proteomic profiles were analyzed via mass spectrometry, comparing newly synthesized "heavy" proteins with pre-existing "light" proteins. RESULTS GDF6 treatment altered protein synthesis in degenerated IVD cells. In NP cells, GDF6 reduced the synthesis of matrisome proteins, including collagens and proteoglycans, while promoting proteins associated with ECM stability, such as LOX, PCOLCE and HAPLN1/3. AF cells demonstrated an upregulation of ECM-stabilizing proteins like POSTN and FMOD. EP cells showed minimal changes, but GDF6 enhanced the synthesis of collagen type II, suggesting improved ECM integrity. Secretome analysis revealed that GDF6 modulated extracellular signalling by promoting ECM-stabilizing proteins and reducing inflammatory markers. CONCLUSION GDF6 exerts compartment-specific effects on protein synthesis in degenerated IVDs, promoting ECM stability, reducing fibrosis, and potentially preserving hydration. These findings support the potential of GDF6 as a therapeutic agent in treating IVD degeneration, particularly in NP-targeted therapies. Future studies should optimize GDF6 dosing and delivery to maximize its regenerative potential.
Collapse
Affiliation(s)
- Vivian Tam
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, Hong Kong S.A.R., China
| | - Neha Chopra
- Spine Labs, St. George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | - Stone Sima
- Spine Labs, St. George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | - Peikai Chen
- Department of Orthopaedics, The University of Hong Kong - Shenzhen Hospital, University of Hong Kong, Shenzhen, China
- The AI and Big Data Lab, The University of Hong Kong - Shenzhen Hospital, Shenzhen, China
| | - Rakesh Sharma
- Proteomics and Metabolomics Core, Centre for PanorOmic Sciences, The University of Hong Kong, Hong Kong, Hong Kong S.A.R., China
| | - Danny Chan
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, Hong Kong S.A.R., China
| | - Ashish Diwan
- Spine Labs, St. George and Sutherland Clinical School, University of New South Wales, Sydney, Australia.
- Spine Service, Department of Orthopaedic Surgery, St. George and Sutherland Clinical School, University of New South Wales, Sydney, Australia.
- Spinal Unit, Discipline of Orthopaedic Surgery, School of Medicine, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
10
|
Wang L, Ruan M, Bu Q, Zhao C. Signaling Pathways Driving MSC Osteogenesis: Mechanisms, Regulation, and Translational Applications. Int J Mol Sci 2025; 26:1311. [PMID: 39941080 PMCID: PMC11818554 DOI: 10.3390/ijms26031311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are crucial for skeletal development, homeostasis, and repair, primarily through their differentiation into osteoblasts and other skeletal lineage cells. Key signaling pathways, including Wnt, TGF-β/BMP, PTH, Hedgehog, and IGF, act as critical regulators of MSC osteogenesis, playing pivotal roles in maintaining bone homeostasis and facilitating regeneration. These pathways interact in distinct ways at various stages of bone development, mineralization, and remodeling. This review provides an overview of the molecular mechanisms by which these pathways regulate MSC osteogenesis, their influence on bone tissue formation, and their implications in bone diseases and therapeutic strategies. Additionally, we explore the potential applications of these pathways in bone tissue engineering, with a particular focus on promoting the use of MSCs as seed cells for bone defect repair. Ultimately, this review aims to highlight potential avenues for advancing bone biology research, treating bone disorders, and enhancing regenerative medicine.
Collapse
Affiliation(s)
| | | | | | - Chengzhu Zhao
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
11
|
Miura T, Watanabe S, Kizaki R, Hasegawa R, Isozaki T, Shimizu M. Production of recombinant masu salmon insulin-like growth factor binding protein-2b1 and its action on pituitary cells. Gen Comp Endocrinol 2025; 363:114674. [PMID: 39914704 DOI: 10.1016/j.ygcen.2025.114674] [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: 07/22/2024] [Revised: 01/19/2025] [Accepted: 02/03/2025] [Indexed: 03/03/2025]
Abstract
Insulin-like growth factor binding protein (Igfbp)-2b is believed to be a major carrier of circulating Igf-1 in salmonids. We cloned cDNAs of two paralogs of igfbp-2b from the liver of masu salmon and produced recombinant Igfbp-2b1 corresponding to the circulating form using a bacterial expression system. The deduced amino acid sequence of masu salmon igfbp-2b1 had a 75.2 % sequence identity with that of masu salmon igfbp-2b2, and 88.7 % and 96.5 % with those of Atlantic salmon and rainbow trout igfbp-2b1, respectively. The coding region of masu salmon igfbp-2b1 cDNA was subcloned into the pET-16b or pET-32a vector and expressed using either a histidine (His)-tag or a thioredoxin (Trx) and His-tag. Recombinant masu salmon (rs) Igfbp-2b1 with the fusion partner was fractionated in the precipitate, solubilized, and isolated using Ni-affinity chromatography. His.rsIgfbp-2b1 and Trx.His.rsIgfbp-2b1 were treated with Factor Xa and enterokinase K, respectively, to remove the fusion partner; only the digestion with enterokinase was successful. After enzymatic digestion, rsIgfbp-2b1 was purified employing reversed-phase high-performance liquid chromatography. The purified rsIgfbp-2b1 was added to a primary culture of masu salmon pituitary cells with or without human (h) IGF-1 to assess its effect on the release of growth hormone (Gh). Although addition of hIGF-1 alone had no effect on Gh release, co-incubation with varying amounts of rsIgfbp-2b1 increased Gh release in a dose-dependent manner. In addition, rsIgfbp-2b1 in the absence of hIGF-1 showed a positive effect on Gh release from salmon pituitary cells. These results suggest that rsIgfbp-2b1 may either have Igf-1-independent action on Gh release or inhibits the suppressive effect of local pituitary Igf-1 on Gh release.
Collapse
Affiliation(s)
- Takuto Miura
- Graduate School of Environmental Science, Hokkaido University, Kita 10, Nishi 5, Kita-ku, Sapporo, Hokkaido 060-0810, Japan
| | - Sota Watanabe
- School of Fisheries Sciences, Hokkaido University, 3-1-1 Minato, Hakodate, Hokkaido 041-8611, Japan
| | - Ryousuke Kizaki
- Graduate School of Fisheries Sciences, Hokkaido University, 3-1-1 Minato, Hakodate, Hokkaido 041-8611, Japan
| | - Ryuya Hasegawa
- Graduate School of Environmental Science, Hokkaido University, Kita 10, Nishi 5, Kita-ku, Sapporo, Hokkaido 060-0810, Japan
| | - Taiga Isozaki
- School of Fisheries Sciences, Hokkaido University, 3-1-1 Minato, Hakodate, Hokkaido 041-8611, Japan
| | - Munetaka Shimizu
- Faculty of Fisheries Sciences, Hokkaido University, 3-1-1 Minato, Hakodate, Hokkaido 041-8611, Japan; Field Science Center for Northern Biosphere, Hokkaido University, 3-1-1 Minato, Hakodate, Hokkaido 041-8611, Japan.
| |
Collapse
|
12
|
Sun Y, Weng X, Chen W, Ge J, Ding B, Ru J, Lei Y, Hu X, Man D, Cheng S, Duan R, Ren J, Yang B. MYBBP1A‑mediated IGFBP4 promoter methylation promotes epithelial‑mesenchymal transition and metastasis through activation of NOTCH pathway in liver cancer. Int J Oncol 2025; 66:4. [PMID: 39611481 PMCID: PMC11637501 DOI: 10.3892/ijo.2024.5710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 11/08/2024] [Indexed: 11/30/2024] Open
Abstract
Metastatic hepatocellular carcinoma (HCC) seriously threatens patients' prognosis. It was previously suggested that the insulin growth factor binding protein (IGFBP) family could serve as cancer suppressors in the development and metastasis of HCC. However, the role of IGFBP4 and its underlying molecular mechanism in HCC metastasis is elusive. In the present study, it was found that IGFBP4 is significantly downregulated in HCC, whose expression is positively correlated with the prognosis of patients with HCC. Overexpression of IGFBP4 restrained migration abilities and cancer metastasis of HCC cells both in vitro and in vivo. Furthermore, it was found that IGFBP4 represses HCC metastasis by inhibiting epithelial‑mesenchymal transition. Molecular mechanism studies showed that overexpression of IGFBP4 obviously suppresses NOTCH1 signaling in HCC. As for the upstream regulatory mechanism, it was revealed that downregulation of IGFBP4 in HCC was caused by CpG islands' hyper‑methylation‑dependent degradation mediated by MYBBP1A. Inhibition of MYBBP1A limited HCC metastatic ability and silence of IGFBP4 at the same time restored HCC metastatic potentials. Clinical data demonstrated that low expression of IGFBP4 was found in patients with HCC, especially with lymphatic metastasis. High MYBBP1A expression and low IGFBP4 expression in HCC were correlated with poor survival of patients with HCC. Summarily, in the present study, it was revealed that MYBBP1A/IGFBP4/NOTCH1 pathway could play a crucial role in the progression and metastasis of HCC, which stimulates novel therapeutic and diagnostic strategies against metastatic HCC.
Collapse
Affiliation(s)
- Yujing Sun
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Xiaoyu Weng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Wei Chen
- General Practice Department, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310005, P.R. China
| | - Jiangzhen Ge
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Bo Ding
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Junnan Ru
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Yunguo Lei
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| | - Xin Hu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P.R. China
| | - Da Man
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Shaobing Cheng
- Department of Colorectal Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Ruoshu Duan
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Jingjing Ren
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Beng Yang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
13
|
Butts B, Kamara J, Morris A, Davis E, Higgins M, Dunbar SB. Comorbid Diabetes Is Associated With Dyspnea Severity and Cardiometabolic Biomarkers in Black Adults With Heart Failure. Nurs Res 2025; 74:27-36. [PMID: 39420458 PMCID: PMC11637969 DOI: 10.1097/nnr.0000000000000784] [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] [Indexed: 10/19/2024]
Abstract
BACKGROUND Comorbidities such as Type 2 diabetes mellitus significantly and adversely influence heart failure outcomes, especially in Black adult populations. Likewise, heart failure has a negative effect on diabetes and cardiometabolic outcomes. Dyspnea, a common symptom of heart failure, often correlates with disease severity and prognosis. However, the relationship between comorbid diabetes, dyspnea severity, and cardiometabolic biomarkers in Black adults with heart failure remains understudied. OBJECTIVES The purpose of this pilot study was to examine differences in the distressing heart failure symptom of dyspnea and in cardiometabolic and inflammatory biomarkers in Black adults living with heart failure with and without diabetes. METHODS Black adults with heart failure were enrolled in this cross-sectional pilot study. Cardiometabolic and inflammatory biomarkers were measured via multiplex immunoassay. Univariate general liner models were used to identify group differences between persons with heart failure with comorbid diabetes and those without, controlling for age, sex, and comorbid burden. RESULTS Participants were mostly female with a mean age of 55 years and mean left ventricular ejection fraction of 33%. Participants with diabetes exhibited higher dyspnea scores compared to those without diabetes, indicating greater symptom burden. Moreover, individuals with comorbid diabetes demonstrated higher levels of cardiometabolic and inflammatory markers. DISCUSSION Comorbid diabetes was associated with higher dyspnea severity and adverse cardiometabolic profiles in Black adults with heart failure. These findings underscore the importance of targeted interventions addressing diabetes management and cardiometabolic risk factors to improve symptom control and outcomes in this high-risk population. Further research is warranted to elucidate the underlying mechanisms and develop tailored therapeutic strategies for managing comorbidities in persons with heart failure, particularly in minoritized communities.
Collapse
Affiliation(s)
- Brittany Butts
- Emory University Nell Hodgson Woodruff School of Nursing, Atlanta, GA
| | - Julia Kamara
- Emory University Nell Hodgson Woodruff School of Nursing, Atlanta, GA
| | | | - Erica Davis
- Emory University Nell Hodgson Woodruff School of Nursing, Atlanta, GA
| | - Melinda Higgins
- Emory University Nell Hodgson Woodruff School of Nursing, Atlanta, GA
| | - Sandra B Dunbar
- Emory University Nell Hodgson Woodruff School of Nursing, Atlanta, GA
| |
Collapse
|
14
|
Khan MZ, Zugaza JL, Torres Aleman I. The signaling landscape of insulin-like growth factor 1. J Biol Chem 2025; 301:108047. [PMID: 39638246 PMCID: PMC11748690 DOI: 10.1016/j.jbc.2024.108047] [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: 07/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
The sheer amplitude of biological actions of insulin-like growth factor I (IGF-1) affecting all types of cells in all tissues suggests a vast signaling landscape for this ubiquitous humoral signal. While the canonical signaling pathways primarily involve the Ras/MAPK and PI3K/AKT cascades, the evolutionary conservation of insulin-like peptides (ILPs) and their pathways hints at the potential for novel functions to emerge over time. Indeed, the evolutionary trajectory of ILPs opens the possibility of either novel functions for these two pathways, novel downstream routes, or both. Evidence supporting this notion includes observations of neofunctionalization in bony fishes or crustaceans, and the involvement of ILPs pathways in invertebrate eusociality or in vertebrate bone physiology, respectively. Such evolutionary processes likely contribute to the rich diversity of ILPs signaling observed today. Moreover, the interplay between conserved signaling pathways, such as those implicated in aging (predominantly involving the PI3K-AKT route), and lesser known pathways, such as those mediated by biased G-protein coupled receptors and others even less known, may underpin the context-dependent actions characteristic of ILPs signaling. While canonical IGF-1 signaling is often assumed to account for the intracellular pathways utilized by this growth factor, a comprehensive analysis of all the pathways mediated by the IGF-1 receptor (IGF-1R) remains lacking. This review aims to explore both canonical and non-canonical routes of IGF-1R action across various cell types, offering a detailed examination of the mechanisms underlying IGF-1 signaling and highlighting the significant gaps in our current understanding.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Achucarro Basque Center for Neuroscience, Leioa, Spain; CIBERNED, Madrid, Spain
| | - Jose Luis Zugaza
- Achucarro Basque Center for Neuroscience, Leioa, Spain; Ikerbasque Science Foundation, Bilbao, Spain
| | - Ignacio Torres Aleman
- Achucarro Basque Center for Neuroscience, Leioa, Spain; CIBERNED, Madrid, Spain; Ikerbasque Science Foundation, Bilbao, Spain.
| |
Collapse
|
15
|
Tong Z, Wu J, Gong Q, Yuan Y, Wang S, Jiang W. Insulin-like growth factor binding protein 7 identified in aged dental pulp by single-cell RNA sequencing. J Adv Res 2024:S2090-1232(24)00596-4. [PMID: 39674503 DOI: 10.1016/j.jare.2024.12.018] [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: 11/23/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024] Open
Abstract
INTRODUCTION Aging influences the regenerative and reparative functions of dental pulp, and an in-depth and complete understanding of aged dental pulp is highly important. OBJECTIVE This study aimed to explore the heterogeneity of young and aged dental pulp tissue via single-cell RNA sequencing (scRNA-seq), search novel markers of aged dental pulp, and further explore their mechanism. METHODS ScRNA-seq was employed to analyze the heterogeneity of young and aged dental pulp tissue, and immunohistochemical staining was used to detect new marker Insulin-like Growth Factor Binding Protein 7 (IGFBP7) in aged dental pulp. Differentially expressed genes (DEGs) between young and aged dental pulp tissue related with senescence-associated secretory phenotype (SASP) were validated in aging model of H2O2-induced dental pulp fibroblast (DPF). The effect of IGFBP7 on cellular senescence were validated by SA-β-Gal, γ-H2AX, and F-actin cytoskeletal staining. RNA-seq was used to analyze the mechanism of IGFBP7 alleviating senescence of H2O2-induced DPFs. RESULTS A total of 32,012 cells were sequenced from 8 dental pulp samples and categorized into 8 main clusters, including fibroblasts (FB), endothelial cells, monocytes, T cells, B cells, mesenchymal stem cells, Schwann cells, and nonmyelinating ScCs. The ratio of fibroblasts was the highest, and FB1 was the largest subcluster of fibroblasts in the young group. In aged dental pulp, the ratio of fibroblasts was relatively low, and fibroblasts had more cellular communication with other cell types in fibroblast growth factor (FGF) and insulin-like growth factor (IGF) signal pathways. IGFBP7 was significantly upregulated in the aged group. Recombinant IGFBP7 reduced the senescence of H2O2-induced DPFs. CONCLUSIONS These findings offer insights into the mechanisms of dental pulp aging and enhance our understanding of dental pulp at the single-cell level. Further comprehensive studies are required to clarify the exact mechanisms through which IGFBP7 influences dental pulp aging.
Collapse
Affiliation(s)
- Zhongchun Tong
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jie Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qimei Gong
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yifang Yuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi 710032, China
| | - Shengchao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi 710032, China.
| | - Wenkai Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
16
|
Hu L, Li D, Wei Q, Kang L, Sun Y, Jiang Y. Characterization of a novel IGFBP-2 transcript in the ovarian granulosa cells of chicken follicles: mRNA expression, function and effect of reproductive hormones and IGF1. Poult Sci 2024; 103:104501. [PMID: 39504834 PMCID: PMC11577207 DOI: 10.1016/j.psj.2024.104501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/11/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024] Open
Abstract
Insulin-like growth factor binding protein-2 (IGFBP-2), a binding protein of insulin-like growth factor (IGF) system, regulates the activity of IGFs and also influences cellular function with endogenous activity. In mammals, IGFBP-2 is reported to affect ovarian follicle development and steroidogenesis; however, its role in the chicken ovary is unknown. In this study, we investigated the mRNA expression and function of a novel IGFBP-2 transcript and the effect of reproductive hormones and insulin-like growth factor 1 (IGF1) on its expression in the ovarian granulosa cells of chicken follicles. The mRNA expression of IGFBP-2 was significantly increased in granulosa cells after follicle selection and was higher in hierarchical granulosa cells (Post-GCs) than in pre-hierarchical granulosa cells (Pre-GCs). IGFBP-2 promoted the proliferation and inhibited the apoptosis of both Pre-GCs and Post-GCs, enhanced the mRNA expression of genes involved in progesterone (P4) synthesis in Pre-GCs. However, in Post-GCs, IGFBP-2 inhibited the mRNA expression of these genes and suppressed P4 secretion. The mRNA expression of IGFBP-2 was inhibited by estradiol (E2) and follicle-stimulating hormone (FSH), but enhanced by P4 in Pre-GCs. In Post-GCs, FSH and IGF1 stimulated the mRNA expression of IGFBP-2 synergistically. Knockdown of IGFBP-2 attenuated the stimulatory effect of IGF1 on the mRNA expression of the side chain cleavage enzyme cytochrome P450 family 11 subfamily A member 1 (CYP11A1). These findings indicate that IGFBP-2 is regulated by FSH and IGF1, exerts different functions in Pre-GCs and Post-GCs in regulating IGF1 and plays an important role in chicken follicle development by affecting granulosa cell proliferation and P4 synthesis.
Collapse
Affiliation(s)
- Longxiao Hu
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271017, China
| | - Dandan Li
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271017, China
| | - Qingqing Wei
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271017, China; Shandong Provincial Key laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian 271017, China
| | - Li Kang
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271017, China; Shandong Provincial Key laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian 271017, China
| | - Yi Sun
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271017, China; Shandong Provincial Key laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian 271017, China
| | - Yunliang Jiang
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271017, China; Shandong Provincial Key laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian 271017, China.
| |
Collapse
|
17
|
Ferreri DM, Sutliffe JT, Lopez NV, Sutliffe CA, Smith R, Carreras-Gallo N, Dwaraka VB, Prestrud AA, Fuhrman JH. Slower Pace of Epigenetic Aging and Lower Inflammatory Indicators in Females Following a Nutrient-Dense, Plant-Rich Diet Than Those in Females Following the Standard American Diet. Curr Dev Nutr 2024; 8:104497. [PMID: 39668946 PMCID: PMC11635705 DOI: 10.1016/j.cdnut.2024.104497] [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: 07/23/2024] [Revised: 10/04/2024] [Accepted: 10/22/2024] [Indexed: 12/14/2024] Open
Abstract
Background Plant-based diets are associated with lower inflammatory biomarkers and reduced risk of age-related chronic diseases. Epigenetic biomarkers of aging are DNA methylation-based tools that estimate biological age and rate of aging, providing insights into age-related health risks. Healthy diet and lifestyle indicators correlate with slower epigenetic aging. Objectives Neither inflammatory biomarkers nor epigenetic aging has yet been studied in the nutrient-dense, plant-rich (Nutritarian) diet, a plant-based diet that emphasizes specific plant foods, such as cruciferous vegetables, beans and other legumes, onions and garlic, mushrooms, berries, nuts, and seeds. We aimed to compare inflammatory status and epigenetic age acceleration in females following a Nutritarian diet with those of females following a standard American diet (SAD). Methods We investigated dietary inflammatory potential, epigenetic age acceleration using first, second, and third-generation clocks, and additional health-related epigenetic biomarkers in this retrospective cohort study of 48 females who habitually (≥5 y) follow a Nutritarian diet and 49 females without obesity who habitually (≥5 y) follow a SAD. Participants completed a series of online questionnaires and provided a blood sample. Results Epigenetic age acceleration, indicated by the third-generation clock DunedinPACE, was significantly slower in the Nutritarian group than that in the SAD group (P = 4.26 × 10-6). The Nutritarian diet group showed lower dietary inflammatory potential, as indicated by Empirical Dietary Inflammatory Pattern and Dietary Inflammatory Index. We observed differences in methylation-predicted immune cell subsets (lower neutrophils and higher T regulatory cells) and a lower epigenetic biomarker proxy for C-reactive protein, both of which suggested a lower inflammatory status in the Nutritarian group. Epigenetic biomarker proxies for LDL cholesterol, body mass index (BMI), insulin-like growth factor binding protein 5, and blood glucose were also lower in the Nutritarian group. Conclusions Our findings suggest the Nutritarian diet could help reduce chronic inflammation and slow epigenetic aging.
Collapse
Affiliation(s)
- Deana M Ferreri
- Nutritional Research Foundation, Flemington, NJ, United States
| | - Jay T Sutliffe
- Department of Health Sciences and the PRANDIAL Lab, Northern Arizona University, Flagstaff, AZ, United States
| | - Nanette V Lopez
- Department of Health Sciences and the PRANDIAL Lab, Northern Arizona University, Flagstaff, AZ, United States
| | - Chloe A Sutliffe
- Department of Health Sciences and the PRANDIAL Lab, Northern Arizona University, Flagstaff, AZ, United States
| | - Ryan Smith
- TruDiagnostic, Lexington, KY, United States
| | | | | | | | - Joel H Fuhrman
- Nutritional Research Foundation, Flemington, NJ, United States
| |
Collapse
|
18
|
王 圆, 来 天, 褚 丹, 白 晶, 严 淑, 秦 海, 郭 瑞. [Megestrol acetate plus metformin for fertility-sparing treatment of atypical endometrial hyperplasia and early-stage endometrial adenocarcinoma: a prospective study]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:2055-2062. [PMID: 39623260 PMCID: PMC11605202 DOI: 10.12122/j.issn.1673-4254.2024.11.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE To evaluate the efficacy of medroxyprogesterone acetate (MA) plus metformin as the primary fertility-sparing treatment for atypical endometrial hyperplasia (AEH) and early-stage grade 1 endometrial adenocarcinoma (G1 EAC) and the recurrence rate after treatment. METHODS Sixty patients (aged 20-42 years) with AEH and/or grade 1 EAC limited to the endometrium were enrolled prospectively and randomized into two groups (n=30) to receive oral MA treatment at the daily dose of 160 mg (control) or MA plus oral metformin (850 mg, twice a day) for at least 6 months. The treatment could extend to 12 months until a complete response (CR) was achieved, and follow-up hysteroscopy and curettage were performed every 3 months. For all the patients who achieved CR, endometrial expressions of IGFBP-rP1, p-Akt and p-AMPK were detected immunohistochemically. RESULTS A total of 58 patients completed the treatment. After 9 months of treatment, 23 (76.7%) patients in the combined treatment group and 20 (71.4%) in the control group achieved CR; two patients in the control group achieved CR after converting to the combined treatment. The recurrence rate did not differ significantly between the control group and combined treatment group (30.0% vs 22.7%, P>0.05). Ten (35.7%) patients in the control group experienced significant weight gain of 5.7±6.1 kg, while none of the patients receiving the combined treatment exhibited significant body weight changes. Compared with the control group, the patients receiving the combined treatment showed enhanced endometrial expressions of IGFBP-rP1 and p-AMPK with lowered p-Akt expression. CONCLUSION Metformin combined with MA may provide an effective option for fertility-sparing treatment of AEH and grade 1 stage IA EAC, and the clinical benefits of metformin for controlling MA-induced weight gain and promoting endometrial expressions of IGFBP-rP1 and p-AMPK while inhibiting p-Akt expression warrants further study.
Collapse
|
19
|
Vaccaro LA, Herring K, Wilson A, England E, Smith AL, Ellestad LE. Dynamic changes in insulin-like growth factor binding protein expression occur between embryonic and early post-hatch development in broiler chickens. Poult Sci 2024; 103:104174. [PMID: 39197338 PMCID: PMC11398635 DOI: 10.1016/j.psj.2024.104174] [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: 02/26/2024] [Revised: 06/07/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024] Open
Abstract
Somatotropic gene expression has been altered by genetic selection, and developmental changes in insulin-like growth factor (IGF) and IGF binding protein (IGFBP) expression may contribute to rapid growth and muscle accretion in commercial broilers. The objective of this study was to evaluate changes in somatotropic axis activity between embryonic day (e) 12 and post-hatch day (d) 21. Liver and breast muscle (pectoralis major) were collected to measure gene expression, and blood was collected post-hatch to measure circulating IGFs. Liver IGF1 rose rapidly post-hatch and, in muscle, IGF1 exhibited a dynamic expression pattern. Levels decreased from e14 to e20, returned to e14 levels at d3, decreased again at d10, and stayed low thereafter. In both tissues, mRNA levels of several IGFBPs changed between embryogenesis and post-hatch. Liver IGFBP2 increased between e12 and e20, returned to e12 levels on d1, and remained low. Conversely, liver IGFBP4 expression was greater post-hatch than during embryogenesis. Expression of select IGFBPs was depressed in liver during the peri-hatch period. Liver IGFBP1, IGFBP3, IGFBP5, and IGFBP7 mRNA levels all decreased around this time and returned to embryonic levels by d3. In breast muscle, expression of both IGFBP2 and IGFBP4 was reduced after hatch. Circulating insulin-like growth factor IGF1 and IGF2 levels did not change between hatch and d21. These data suggest that post-hatch IGF effects are likely modulated by target tissue IGFR1 and IGFBP expression rather than changes in circulating hormone levels, with promotion or restriction of IGF-receptor binding regulating growth. Downregulation of several IGFBPs synthesized in the liver may facilitate the metabolic transition from utilizing yolk lipids to dietary carbohydrates. Several IGFBPs produced in breast muscle appear to have growth-promotive effects during embryogenesis but restrict growth of this tissue after hatch, as their post-hatch downregulation could facilitate local IGF signaling. These developmental gene expression patterns suggest that somatotropic hormonal signaling regulating growth and muscle accretion might be controlled through differential actions of IGFBPs and provide a basis for future functional studies.
Collapse
Affiliation(s)
- Lauren A Vaccaro
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Kyle Herring
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Abigail Wilson
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Emma England
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Addison L Smith
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Laura E Ellestad
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
20
|
Wang M, Wang H, Wang X, Shen Y, Zhou D, Jiang Y. Identification of cellular senescence-related genes and immune cell infiltration characteristics in intervertebral disc degeneration. Front Immunol 2024; 15:1439976. [PMID: 39328407 PMCID: PMC11424418 DOI: 10.3389/fimmu.2024.1439976] [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: 05/29/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Background Intervertebral disc degeneration (IDD) progression involves multiple factors, including loss of nucleus pulposus cells and extracellular matrix as the basic pathological mechanism of degeneration, and is closely related to cellular senescence and immune cell infiltration. The aim of study was to identify critical cellular senescence-related genes and immune cell infiltration characteristics in IDD. Methods Four datasets, including GSE70362, GSE112216, GSE114169, and GSE150408, were downloaded from the Gene Expression Omnibus database. The senescence-related genes were acquired from the CellAge Database and intersected with differentially expressed genes (DEGs) between IDD and control samples for senescence-related DEGs (SRDEGs). Protein-protein interaction (PPI) network analysis was performed to obtain ten hub SRDEGs. A consensus cluster analysis based on these hub genes was performed to divide the patients into clusters. The functional enrichment, and immune infiltration statuses of the clusters were compared. Weighted gene co-expression network analysis was used to identified key gene modules. The overlapping genes from key modules, DEGs of clusters and hub SRDEGs were intersected to obtain potential biomarkers. To verify the expression of potential biomarkers, quantitative polymerase chain reaction (qPCR) and immunohistochemistry were performed by using human intervertebral disc tissues. Results In the GSE70362 dataset, a total of 364 DEGs were identified, of which 150 were upregulated and 214 were downregulated, and 35 genes were selected as SRDEGs. PPI analysis revealed ten hub SRDEGs and consensus cluster analysis divided the patients into two clusters. Compared to Cluster 2, Cluster 1 was highly enriched in extracellular matrix organization and various metabolic process. The level of Follicular T helper cells in the Cluster 1 was significantly higher than that in the Cluster 2. IGFBP3 and NQO1 were identified as potential biomarkers. The remaining 3 datasets, and the result of qPCR and immunohistochemistry showed that the expression levels of NQO1 and IGFBP3 in the degenerated group were higher than those in the control or treatment groups. Conclusion Senescence-related genes play a key role in the development and occurrence of IDD. IGFBP3 and NQO1 are strongly correlated with immune infiltration in the IDD and could become novel therapeutic targets that prevent the progression of IDD.
Collapse
Affiliation(s)
- Muyi Wang
- Department of Orthopedics, Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Hao Wang
- Department of Orthopedics, Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Xin Wang
- Department of Orthopedics, Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yifei Shen
- Department of Orthopedics, Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Dong Zhou
- Department of Orthopedics, Affiliated Changzhou Children’s Hospital of Nantong University, Changzhou, Jiangsu, China
| | - Yuqing Jiang
- Department of Orthopedics, Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
21
|
Li X, Lin S, Yang X, Chen C, Cao S, Zhang Q, Ma J, Zhu G, Zhang Q, Fang Q, Zheng C, Liang W, Wu X. When IGF-1 Meets Metabolic Inflammation and Polycystic Ovary Syndrome. Int Immunopharmacol 2024; 138:112529. [PMID: 38941670 DOI: 10.1016/j.intimp.2024.112529] [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: 04/12/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder associated with insulin resistance (IR) and hyperandrogenaemia (HA). Metabolic inflammation (MI), characterized by a chronic low-grade inflammatory state, is intimately linked with chronic metabolic diseases such as IR and diabetes and is also considered an essential factor in the development of PCOS. Insulin-like growth factor 1 (IGF-1) plays an essential role in PCOS pathogenesis through its multiple functions in regulating cell proliferation metabolic processes and reducing inflammatory responses. This review summarizes the molecular mechanisms by which IGF-1, via MI, participates in the onset and progression of PCOS, aiming to provide insights for studies and clinical treatment of PCOS.
Collapse
Affiliation(s)
- Xiushen Li
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China; Department of Traditional Chinese Medicine, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Sailing Lin
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Xiaolu Yang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Can Chen
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Shu Cao
- Xin'an Academy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Qi Zhang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Jingxin Ma
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Guli Zhu
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Qi Zhang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Qiongfang Fang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Weizheng Liang
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China.
| | - Xueqing Wu
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China.
| |
Collapse
|
22
|
Wang X, Cao L, Liu S, Zhou Y, Zhou J, Zhao W, Gao S, Liu R, Shi Y, Shao C, Fang J. The critical roles of IGFs in immune modulation and inflammation. Cytokine 2024; 183:156750. [PMID: 39243567 DOI: 10.1016/j.cyto.2024.156750] [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: 06/03/2024] [Revised: 07/31/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Insulin-like growth factors (IGFs) are crucial for embryonic and postnatal growth and development, influencing cell survival, metabolism, myogenesis, and cancer progression. Many studies have demonstrated that IGFs also play prominent roles in the modulation of both innate and adaptive immune systems during inflammation. Strikingly, IGFs dictate the phenotype and functional properties of macrophages and T cells. Furthermore, the interplay between IGFs and inflammatory cytokines may generate tissue-protective properties during inflammation. Herein, we review the recent advances on the dialogue between immune cells and IGFs, especially zooming in on the significance of immunomodulatory properties in inflammatory conditions, cancer and autoimmune diseases. The investigation of IGFs may have broad clinical implications.
Collapse
Affiliation(s)
- Xin Wang
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Lijuan Cao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China; Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Shisong Liu
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yipeng Zhou
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jiarui Zhou
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wenxuan Zhao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Shengqi Gao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Rui Liu
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China; Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Yufang Shi
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China; Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Changshun Shao
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Jiankai Fang
- The Third/Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
23
|
Matuszewska J, Krawiec A, Radziemski A, Uruski P, Tykarski A, Mikuła-Pietrasik J, Książek K. Alterations of receptors and insulin-like growth factor binding proteins in senescent cells. Eur J Cell Biol 2024; 103:151438. [PMID: 38945074 DOI: 10.1016/j.ejcb.2024.151438] [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: 02/08/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024] Open
Abstract
The knowledge about cellular senescence expands dynamically, providing more and more conclusive evidence of its triggers, mechanisms, and consequences. Senescence-associated secretory phenotype (SASP), one of the most important functional traits of senescent cells, is responsible for a large extent of their context-dependent activity. Both SASP's components and signaling pathways are well-defined. A literature review shows, however, that a relatively underinvestigated aspect of senescent cell autocrine and paracrine activity is the change in the production of proteins responsible for the reception and transmission of SASP signals, i.e., receptors and binding proteins. For this reason, we present in this article the current state of knowledge regarding senescence-associated changes in cellular receptors and insulin-like growth factor binding proteins. We also discuss the role of these alterations in senescence induction and maintenance, pro-cancerogenic effects of senescent cells, and aging-related structural and functional malfunctions.
Collapse
Affiliation(s)
- Julia Matuszewska
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland
| | - Adrianna Krawiec
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland
| | - Artur Radziemski
- Poznan University of Medical Sciences, Department of Hypertensiology, Długa 1/2 Str., Poznań 61-848, Poland
| | - Paweł Uruski
- Poznan University of Medical Sciences, Department of Hypertensiology, Długa 1/2 Str., Poznań 61-848, Poland
| | - Andrzej Tykarski
- Poznan University of Medical Sciences, Department of Hypertensiology, Długa 1/2 Str., Poznań 61-848, Poland
| | - Justyna Mikuła-Pietrasik
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland
| | - Krzysztof Książek
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland.
| |
Collapse
|
24
|
Kotsifaki A, Maroulaki S, Karalexis E, Stathaki M, Armakolas A. Decoding the Role of Insulin-like Growth Factor 1 and Its Isoforms in Breast Cancer. Int J Mol Sci 2024; 25:9302. [PMID: 39273251 PMCID: PMC11394947 DOI: 10.3390/ijms25179302] [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: 07/30/2024] [Revised: 08/25/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
Insulin-like Growth Factor-1 (IGF-1) is a crucial mitogenic factor with important functions in the mammary gland, mainly through its interaction with the IGF-1 receptor (IGF-1R). This interaction activates a complex signaling network that promotes cell proliferation, epithelial to mesenchymal transition (EMT) and inhibits apoptosis. Despite extensive research, the precise molecular pathways and intracellular mechanisms activated by IGF-1, in cancer, remain poorly understood. Recent evidence highlights the essential roles of IGF-1 and its isoforms in breast cancer (BC) development, progression, and metastasis. The peptides that define the IGF-1 isoforms-IGF-1Ea, IGF-1Eb, and IGF-1Ec-act as key points of convergence for various signaling pathways that influence the growth, metastasis and survival of BC cells. The aim of this review is to provide a detailed exami-nation of the role of the mature IGF-1 and its isoforms in BC biology and their potential use as possible therapeutical targets.
Collapse
Affiliation(s)
- Amalia Kotsifaki
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Sousanna Maroulaki
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Efthymios Karalexis
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Martha Stathaki
- Surgical Clinic, "Elena Venizelou" General Hospital, 11521 Athens, Greece
| | - Athanasios Armakolas
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
25
|
Chen L, Hui L, Li J. The multifaceted role of insulin-like growth factor binding protein 7. Front Cell Dev Biol 2024; 12:1420862. [PMID: 39081862 PMCID: PMC11286461 DOI: 10.3389/fcell.2024.1420862] [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: 04/24/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Insulin-like growth factor binding protein 7 (IGFBP7) serves as a crucial extracellular matrix protein, exerting pivotal roles in both physiological and pathological processes. This comprehensive review meticulously delineates the structural attributes of IGFBP7, juxtaposing them with other members within the IGFBP families, and delves into the expression patterns across various tissues. Furthermore, the review thoroughly examines the multifaceted functions of IGFBP7, encompassing its regulatory effects on cell proliferation, apoptosis, and migration, elucidating the underlying mechanistic pathways. Moreover, it underscores the compelling roles in tumor progression, acute kidney injury, and reproductive processes. By rigorously elucidating the diverse functionalities and regulatory networks of IGFBP7 across various physiological and pathological contexts, this review aims to furnish a robust theoretical framework and delineate future research trajectories for leveraging IGFBP7 in disease diagnosis, therapeutic interventions, and pharmaceutical innovations.
Collapse
Affiliation(s)
| | | | - Jun Li
- Department of Immunology, Center of Immunomolecular Engineering, Innovation and Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
26
|
Scheifler M, Wilhelm L, Visser B. Lipid Metabolism in Parasitoids and Parasitized Hosts. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 38977639 DOI: 10.1007/5584_2024_812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Parasitoids have an exceptional lifestyle where juvenile development is spent on or in a single host insect, but the adults are free-living. Unlike parasites, parasitoids kill the host. How parasitoids use such a limiting resource, particularly lipids, can affect chances to survive and reproduce. In part 1, we describe the parasitoid lifestyle, including typical developmental strategies. Lipid metabolism in parasitoids has been of interest to researchers since the 1960s and continues to fascinate ecologists, evolutionists, physiologists, and entomologists alike. One reason of this interest is that the majority of parasitoids do not accumulate triacylglycerols as adults. Early research revealed that some parasitoid larvae mimic the fatty acid composition of the host, which may result from a lack of de novo triacylglycerol synthesis. More recent work has focused on the evolution of lack of adult triacylglycerol accumulation and consequences for life history traits. In part 2 of this chapter, we discuss research efforts on lipid metabolism in parasitoids from the 1960s onwards. Parasitoids are also master manipulators of host physiology, including lipid metabolism, having evolved a range of mechanisms to affect the release, synthesis, transport, and take-up of lipids from the host. We lay out the effects of parasitism on host physiology in part 3 of this chapter.
Collapse
Affiliation(s)
- Mathilde Scheifler
- Evolution and Ecophysiology Group, Department of Functional and Evolutionary Entomology, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Léonore Wilhelm
- Evolution and Ecophysiology Group, Department of Functional and Evolutionary Entomology, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Bertanne Visser
- Evolution and Ecophysiology Group, Department of Functional and Evolutionary Entomology, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium.
| |
Collapse
|
27
|
Baxter RC. Endocrine and cellular physiology and pathology of the insulin-like growth factor acid-labile subunit. Nat Rev Endocrinol 2024; 20:414-425. [PMID: 38514815 DOI: 10.1038/s41574-024-00970-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 03/23/2024]
Abstract
The acid-labile subunit (ALS) of the insulin-like growth factor (IGF) binding protein (IGFBP) complex, encoded in humans by IGFALS, has a vital role in regulating the endocrine transport and bioavailability of IGF-1 and IGF-2. Accordingly, ALS has a considerable influence on postnatal growth and metabolism. ALS is a leucine-rich glycoprotein that forms high-affinity ternary complexes with IGFBP-3 or IGFBP-5 when they are occupied by either IGF-1 or IGF-2. These complexes constitute a stable reservoir of circulating IGFs, blocking the potentially hypoglycaemic activity of unbound IGFs. ALS is primarily synthesized by hepatocytes and its expression is lower in non-hepatic tissues. ALS synthesis is strongly induced by growth hormone and suppressed by IL-1β, thus potentially serving as a marker of growth hormone secretion and/or activity and of inflammation. IGFALS mutations in humans and Igfals deletion in mice cause modest growth retardation and pubertal delay, accompanied by decreased osteogenesis and enhanced adipogenesis. In hepatocellular carcinoma, IGFALS is described as a tumour suppressor; however, its contribution to other cancers is not well delineated. This Review addresses the endocrine physiology and pathology of ALS, discusses the latest cell and proteomic studies that suggest emerging cellular roles for ALS and outlines its involvement in other disease states.
Collapse
Affiliation(s)
- Robert C Baxter
- University of Sydney, Kolling Institute, Royal North Shore Hospital, St Leonards, New South Wales, Australia.
| |
Collapse
|
28
|
Pye D, Scholey R, Ung S, Dawson M, Shahmalak A, Purba TS. Activation of the integrated stress response in human hair follicles. PLoS One 2024; 19:e0303742. [PMID: 38900734 PMCID: PMC11189182 DOI: 10.1371/journal.pone.0303742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/30/2024] [Indexed: 06/22/2024] Open
Abstract
Unravelling how energy metabolism and stress responses are regulated in human scalp hair follicles could reveal novel insights into the controls of hair growth and provide new targets to manage hair loss disorders. The Mitochondrial Pyruvate Carrier (MPC) imports pyruvate, produced via glycolysis, into the mitochondria, fuelling the TCA cycle. Previous work has shown that MPC inhibition promotes lactate generation, which activates murine epithelial hair follicle stem cells (eHFSCs). However, by pharmacologically targeting the MPC in short-term human hair follicle ex vivo organ culture experiments using UK-5099, we induced metabolic stress-responsive proliferative arrest throughout the human hair follicle epithelium, including within Keratin 15+ eHFSCs. Through transcriptomics, MPC inhibition was shown to promote a gene expression signature indicative of disrupted FGF, IGF, TGFβ and WNT signalling, mitochondrial dysfunction, and activation of the integrated stress response (ISR), which can arrest cell cycle progression. The ISR, mediated by the transcription factor ATF4, is activated by stressors including amino acid deprivation and ER stress, consistent with MPC inhibition within our model. Using RNAScope, we confirmed the upregulation of both ATF4 and the highly upregulated ATF4-target gene ADM2 on human hair follicle tissue sections in situ. Moreover, treatment with the ISR inhibitor ISRIB attenuated both the upregulation of ADM2 and the proliferative block imposed via MPC inhibition. Together, this work reveals how the human hair follicle, as a complex and metabolically active human tissue system, can dynamically adapt to metabolic stress.
Collapse
Affiliation(s)
- Derek Pye
- Division Musculoskeletal and Dermatological Sciences, Centre for Dermatology Research, Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, School of Biosciences, The University of Manchester, Manchester, United Kingdom
| | - Rachel Scholey
- Bioinformatics Core Facility, University of Manchester, Manchester, United Kingdom
| | - Sin Ung
- Division Musculoskeletal and Dermatological Sciences, Centre for Dermatology Research, Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, School of Biosciences, The University of Manchester, Manchester, United Kingdom
| | - Madoc Dawson
- Division Musculoskeletal and Dermatological Sciences, Centre for Dermatology Research, Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, School of Biosciences, The University of Manchester, Manchester, United Kingdom
| | | | - Talveen S. Purba
- Division Musculoskeletal and Dermatological Sciences, Centre for Dermatology Research, Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, School of Biosciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
29
|
Schlueter BC, Quanz K, Baldauf J, Petrovic A, Ruppert C, Guenther A, Gall H, Tello K, Grimminger F, Ghofrani HA, Weissmann N, Seeger W, Schermuly RT, Weiss A. The diverging roles of insulin-like growth factor binding proteins in pulmonary arterial hypertension. Vascul Pharmacol 2024; 155:107379. [PMID: 38762131 DOI: 10.1016/j.vph.2024.107379] [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: 06/29/2023] [Revised: 12/29/2023] [Accepted: 05/05/2024] [Indexed: 05/20/2024]
Abstract
Pulmonary hypertension (PH) is a progressive, severe and to date not curable disease of the pulmonary vasculature. Alterations of the insulin-like growth factor 1 (IGF-1) system are known to play a role in vascular pathologies and IGF-binding proteins (IGFBPs) are important regulators of the bioavailability and function of IGFs. In this study, we show that circulating plasma levels of IGFBP-1, IGFBP-2 and IGFBP-3 are increased in idiopathic pulmonary arterial hypertension (IPAH) patients compared to healthy individuals. These binding proteins inhibit the IGF-1 induced IGF-1 receptor (IGF1R) phosphorylation and exhibit diverging effects on the IGF-1 induced signaling pathways in human pulmonary arterial cells (i.e. healthy as well as IPAH-hPASMCs, and healthy hPAECs). Furthermore, IGFBPs are differentially expressed in an experimental mouse model of PH. In hypoxic mouse lungs, IGFBP-1 mRNA expression is decreased whereas the mRNA for IGFBP-2 is increased. In contrast to IGFBP-1, IGFBP-2 shows vaso-constrictive properties in the murine pulmonary vasculature. Our analyses show that IGFBP-1 and IGFBP-2 exhibit diverging effects on IGF-1 signaling and display a unique IGF1R-independent kinase activation pattern in human pulmonary arterial smooth muscle cells (hPASMCs), which represent a major contributor of PAH pathobiology. Furthermore, we could show that IGFBP-2, in contrast to IGFBP-1, induces epidermal growth factor receptor (EGFR) signaling, Stat-3 activation and expression of Stat-3 target genes. Based on our results, we conclude that the IGFBP family, especially IGFBP-1, IGFBP-2 and IGFBP-3, are deregulated in PAH, that they affect IGF signaling and thereby regulate the cellular phenotype in PH.
Collapse
MESH Headings
- Humans
- Animals
- Receptor, IGF Type 1/metabolism
- Receptor, IGF Type 1/genetics
- Signal Transduction
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Insulin-Like Growth Factor Binding Protein 3/metabolism
- Insulin-Like Growth Factor Binding Protein 3/genetics
- Insulin-Like Growth Factor Binding Protein 2/metabolism
- Insulin-Like Growth Factor Binding Protein 2/genetics
- Insulin-Like Growth Factor I/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Cells, Cultured
- Male
- Insulin-Like Growth Factor Binding Protein 1/metabolism
- Insulin-Like Growth Factor Binding Protein 1/genetics
- Phosphorylation
- Disease Models, Animal
- STAT3 Transcription Factor/metabolism
- Case-Control Studies
- Mice, Inbred C57BL
- Familial Primary Pulmonary Hypertension/metabolism
- Familial Primary Pulmonary Hypertension/physiopathology
- Familial Primary Pulmonary Hypertension/pathology
- Familial Primary Pulmonary Hypertension/genetics
- Female
- ErbB Receptors/metabolism
- Middle Aged
- Vascular Remodeling
- Adult
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
Collapse
Affiliation(s)
- Beate Christiane Schlueter
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany
| | - Karin Quanz
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany
| | - Julia Baldauf
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany
| | - Aleksandar Petrovic
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany
| | - Clemens Ruppert
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany
| | - Andreas Guenther
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany; Agaplesion Lung Clinic Waldhof-Elgershausen, Greifenstein 35753, Germany
| | - Henning Gall
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany; University Hospital Giessen and Marburg (UKGM), Giessen 35392, Germany
| | - Khodr Tello
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany; University Hospital Giessen and Marburg (UKGM), Giessen 35392, Germany
| | - Friedrich Grimminger
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany; University Hospital Giessen and Marburg (UKGM), Giessen 35392, Germany
| | - Hossein-Ardeschir Ghofrani
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany; University Hospital Giessen and Marburg (UKGM), Giessen 35392, Germany
| | - Norbert Weissmann
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany
| | - Werner Seeger
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany; Max Planck Institute (MPI) for Heart and Lung Research, Parkstrasse 1, Bad Nauheim 61231, Germany; University Hospital Giessen and Marburg (UKGM), Giessen 35392, Germany
| | - Ralph Theo Schermuly
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany
| | - Astrid Weiss
- Justus-Liebig-University Giessen (JLU), Aulweg 130, Giessen 35392, Germany; Universities of Giessen and Marburg Lung Center (UGMLC), Giessen 35392, Germany; Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Giessen 35392, Germany; Member of the German Center for Lung Research (DZL), Giessen 35392, Germany.
| |
Collapse
|
30
|
Elemam NM, Hotait HY, Saleh MA, El-Huneidi W, Talaat IM. Insulin-like growth factor family and prostate cancer: new insights and emerging opportunities. Front Endocrinol (Lausanne) 2024; 15:1396192. [PMID: 38872970 PMCID: PMC11169579 DOI: 10.3389/fendo.2024.1396192] [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: 03/05/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024] Open
Abstract
Prostate cancer is the second most commonly diagnosed cancer in men. The mammalian insulin-like growth factor (IGF) family is made up of three ligands (IGF-I, IGF-II, and insulin), three receptors (IGF-I receptor (IGF-1R), insulin receptor (IR), and IGF-II receptor (IGF-2R)), and six IGF-binding proteins (IGFBPs). IGF-I and IGF-II were identified as potent mitogens and were previously associated with an increased risk of cancer development including prostate cancer. Several reports showed controversy about the expression of the IGF family and their connection to prostate cancer risk due to the high degree of heterogeneity among prostate tumors, sampling bias, and evaluation techniques. Despite that, it is clear that several IGF family members play a role in prostate cancer development, metastasis, and androgen-independent progression. In this review, we aim to expand our understanding of prostate tumorigenesis and regulation through the IGF system. Further understanding of the role of IGF signaling in PCa shows promise and needs to be considered in the context of a comprehensive treatment strategy.
Collapse
Affiliation(s)
- Noha M. Elemam
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Mohamed A. Saleh
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Waseem El-Huneidi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Iman M. Talaat
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
31
|
Gong J, Ding G, Hao Z, Li Y, Deng A, Zhang C. Elucidating the mechanism of corneal epithelial cell repair: unraveling the impact of growth factors. Front Med (Lausanne) 2024; 11:1384500. [PMID: 38638937 PMCID: PMC11024251 DOI: 10.3389/fmed.2024.1384500] [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: 02/09/2024] [Accepted: 03/21/2024] [Indexed: 04/20/2024] Open
Abstract
The repair mechanism for corneal epithelial cell injuries encompasses migration, proliferation, and differentiation of corneal epithelial cells, and extracellular matrix remodeling of the stromal structural integrity. Furthermore, it involves the consequential impact of corneal limbal stem cells (LSCs). In recent years, as our comprehension of the mediating mechanisms underlying corneal epithelial injury repair has advanced, it has become increasingly apparent that growth factors play a pivotal role in this intricate process. These growth factors actively contribute to the restoration of corneal epithelial injuries by orchestrating responses and facilitating specific interactions at targeted sites. This article systematically summarizes the role of growth factors in corneal epithelial cell injury repair by searching relevant literature in recent years, and explores the limitations of current literature search, providing a certain scientific basis for subsequent basic research and clinical applications.
Collapse
Affiliation(s)
- Jinjin Gong
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Ophthalmology, Jinan Second People’s Hospital, Jinan, China
| | - Gang Ding
- Department of Ophthalmology, Jinan Second People’s Hospital, Jinan, China
| | - Zhongkai Hao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Ophthalmology, Jinan Second People’s Hospital, Jinan, China
| | - Yuchun Li
- Wuxi No. 2 Chinese Medicine Hospital, Wuxi, China
| | - Aijun Deng
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Chenming Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Ophthalmology, Jinan Second People’s Hospital, Jinan, China
| |
Collapse
|
32
|
Lee J, Huh S, Park K, Kang N, Yu HS, Park HG, Kim YS, Kang UG, Won S, Kim SH. Behavioral and transcriptional effects of repeated electroconvulsive seizures in the neonatal MK-801-treated rat model of schizophrenia. Psychopharmacology (Berl) 2024; 241:817-832. [PMID: 38081977 DOI: 10.1007/s00213-023-06511-7] [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: 10/23/2023] [Accepted: 11/23/2023] [Indexed: 03/13/2024]
Abstract
RATIONALE Electroconvulsive therapy (ECT) is an effective treatment modality for schizophrenia. However, its antipsychotic-like mechanism remains unclear. OBJECTIVES To gain insight into the antipsychotic-like actions of ECT, this study investigated how repeated treatments of electroconvulsive seizure (ECS), an animal model for ECT, affect the behavioral and transcriptomic profile of a neurodevelopmental animal model of schizophrenia. METHODS Two injections of MK-801 or saline were administered to rats on postnatal day 7 (PN7), and either repeated ECS treatments (E10X) or sham shock was conducted daily from PN50 to PN59. Ultimately, the rats were divided into vehicle/sham (V/S), MK-801/sham (M/S), vehicle/ECS (V/E), and MK-801/ECS (M/E) groups. On PN59, prepulse inhibition and locomotor activity were tested. Prefrontal cortex transcriptomes were analyzed with mRNA sequencing and network and pathway analyses, and quantitative real-time polymerase chain reaction (qPCR) analyses were subsequently conducted. RESULTS Prepulse inhibition deficit was induced by MK-801 and normalized by E10X. In M/S vs. M/E model, Egr1, Mmp9, and S100a6 were identified as center genes, and interleukin-17 (IL-17), nuclear factor kappa B (NF-κB), and tumor necrosis factor (TNF) signaling pathways were identified as the three most relevant pathways. In the V/E vs. V/S model, mitophagy, NF-κB, and receptor for advanced glycation end products (RAGE) pathways were identified. qPCR analyses demonstrated that Igfbp6, Btf3, Cox6a2, and H2az1 were downregulated in M/S and upregulated in M/E. CONCLUSIONS E10X reverses the behavioral changes induced by MK-801 and produces transcriptional changes in inflammatory, insulin, and mitophagy pathways, which provide mechanistic insight into the antipsychotic-like mechanism of ECT.
Collapse
Affiliation(s)
- Jeonghoon Lee
- Department of Psychiatry, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seonghoo Huh
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyungtaek Park
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - Nuree Kang
- Department of Psychiatry, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Sook Yu
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hong Geun Park
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yong Sik Kim
- Department of Psychiatry, Nowon Eulji Medical Center, Eulji University, Seoul, Republic of Korea
| | - Ung Gu Kang
- Department of Psychiatry, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human Behavioral Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sungho Won
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
- Interdisciplinary Program of Bioinformatics, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- RexSoft Inc., Seoul, Republic of Korea
| | - Se Hyun Kim
- Department of Psychiatry, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
33
|
Iqbal A, Van Hul N, Belicova L, Corbat AA, Hankeova S, Andersson ER. Spatially segregated defects and IGF1-responsiveness of hilar and peripheral biliary organoids from a model of Alagille syndrome. Liver Int 2024; 44:541-558. [PMID: 38014627 DOI: 10.1111/liv.15789] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND & AIMS Alagille syndrome (ALGS) manifests with peripheral intrahepatic bile duct (IHBD) paucity, which can spontaneously resolve. In a model for ALGS, Jag1Ndr/Ndr mice, this occurs with distinct architectural mechanisms in hilar and peripheral IHBDs. Here, we investigated region-specific IHBD characteristics and addressed whether IGF1, a cholangiocyte mitogen that is downregulated in ALGS and in Jag1Ndr/Ndr mice, can improve biliary outcomes. METHODS Intrahepatic cholangiocyte organoids (ICOs) were derived from hilar and peripheral adult Jag1+/+ and Jag1Ndr/Ndr livers (hICOs and pICOs, respectively). ICOs were grown in Matrigel or microwell arrays, and characterized using bulk RNA sequencing, immunofluorescence, and high throughput analyses of nuclear sizes. ICOs were treated with IGF1, followed by analyses of growth, proliferation, and death. CellProfiler and Python scripts were custom written for image analyses. Key results were validated in vivo by immunostaining. RESULTS Cell growth assays and transcriptomics demonstrated that Jag1Ndr/Ndr ICOs were less proliferative than Jag1+/+ ICOs. IGF1 specifically rescued survival and growth of Jag1Ndr/Ndr pICOs. Jag1Ndr/Ndr hICOs were the least proliferative, with lower Notch signalling and an enrichment of hepatocyte signatures and IGF uptake/transport pathways. In vitro (Jag1Ndr/Ndr hICOs) and in vivo (Jag1Ndr/Ndr hilar portal tracts) analyses revealed ectopic HNF4a+ hepatocytes. CONCLUSIONS Hilar and peripheral Jag1Ndr/Ndr ICOs exhibit differences in Notch signalling status, proliferation, and cholangiocyte commitment which may result in cholangiocyte-to-hepatocyte transdifferentiation. While Jag1Ndr/Ndr pICOs can be rescued by IGF1, hICOs are unresponsive, perhaps due to their hepatocyte-like state and/or expression of IGF transport components. IGF1 represents a potential therapeutic for peripheral bile ducts.
Collapse
Affiliation(s)
- Afshan Iqbal
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Noemi Van Hul
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Lenka Belicova
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Agustin A Corbat
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Simona Hankeova
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Emma R Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Hu Z, Zhao X, Wu Z, Qu B, Yuan M, Xing Y, Song Y, Wang Z. Lymphatic vessel: origin, heterogeneity, biological functions, and therapeutic targets. Signal Transduct Target Ther 2024; 9:9. [PMID: 38172098 PMCID: PMC10764842 DOI: 10.1038/s41392-023-01723-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 11/03/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
Lymphatic vessels, comprising the secondary circulatory system in human body, play a multifaceted role in maintaining homeostasis among various tissues and organs. They are tasked with a serious of responsibilities, including the regulation of lymph absorption and transport, the orchestration of immune surveillance and responses. Lymphatic vessel development undergoes a series of sophisticated regulatory signaling pathways governing heterogeneous-origin cell populations stepwise to assemble into the highly specialized lymphatic vessel networks. Lymphangiogenesis, as defined by new lymphatic vessels sprouting from preexisting lymphatic vessels/embryonic veins, is the main developmental mechanism underlying the formation and expansion of lymphatic vessel networks in an embryo. However, abnormal lymphangiogenesis could be observed in many pathological conditions and has a close relationship with the development and progression of various diseases. Mechanistic studies have revealed a set of lymphangiogenic factors and cascades that may serve as the potential targets for regulating abnormal lymphangiogenesis, to further modulate the progression of diseases. Actually, an increasing number of clinical trials have demonstrated the promising interventions and showed the feasibility of currently available treatments for future clinical translation. Targeting lymphangiogenic promoters or inhibitors not only directly regulates abnormal lymphangiogenesis, but improves the efficacy of diverse treatments. In conclusion, we present a comprehensive overview of lymphatic vessel development and physiological functions, and describe the critical involvement of abnormal lymphangiogenesis in multiple diseases. Moreover, we summarize the targeting therapeutic values of abnormal lymphangiogenesis, providing novel perspectives for treatment strategy of multiple human diseases.
Collapse
Affiliation(s)
- Zhaoliang Hu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Xushi Zhao
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Zhonghua Wu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Bicheng Qu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Minxian Yuan
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Yanan Xing
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
| | - Yongxi Song
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
| |
Collapse
|
35
|
Perks CM. Role of the Insulin-like Growth Factor (IGF) Axis in Diseases. Int J Mol Sci 2023; 24:16969. [PMID: 38069291 PMCID: PMC10706945 DOI: 10.3390/ijms242316969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
The insulin-like growth factor axis is a multifaceted, complex system that comprises two ligands, IGF-I and IGF-II, receptors (IGF-1R, IGF-IIR, insulin receptor isoforms IR-A and B, and hybrid receptors) six high affinity IGF-binding proteins (IGFBPs 1-6), and IGFBP proteases [...].
Collapse
Affiliation(s)
- Claire M Perks
- Cancer Endocrinology Group, Bristol Medical School, Translational Health Sciences, Learning & Research Building, Southmead Hospital, Bristol BS105NB, UK
| |
Collapse
|
36
|
Gui R, Li W, Li Z, Wang H, Wu Y, Jiao W, Zhao G, Shen Y, Wang L, Zhang J, Chen S, Hao L, Cheng Y. Effects and potential mechanisms of IGF1/IGF1R in the liver fibrosis: A review. Int J Biol Macromol 2023; 251:126263. [PMID: 37567540 DOI: 10.1016/j.ijbiomac.2023.126263] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Liver fibrosis is a wound-healing response due to persistent liver damage and it may progress to cirrhosis and even liver cancer if no intervention is given. In the current cognition, liver fibrosis is reversible. So, it is of great significance to explore the related gene targets or biomarker for anti-fibrosis of liver. Insulin like growth factor 1 (IGF1) and IGF1 receptor (IGF1R) are mainly expressed in the liver tissues and play critical roles in the liver function. The present review summarized the role of IGF1/IGF1R and its signaling system in liver fibrosis and illustrated the potential mechanisms including DNA damage repair, cell senescence, lipid metabolism and oxidative stress that may be involved in this process according to the studies on the fibrosis of liver or other organs. In particular, the roles of IGF1 and IGF1R in DNA damage repair were elaborated, including membrane-localized and nucleus-localized IGF1R. In addition, for each of the potential mechanism in anti-fibrosis of liver, the signaling pathways of the IGF1/IGF1R mediated and the cell species in liver acted by IGF1 and IGF1R under different conditions were included. The data in this review will support for the study about the effect of IGF1/IGF1R on liver fibrosis induced by various factors, meanwhile, provide a basis for the study of liver fibrosis to focus on the communications between the different kinds of liver cells.
Collapse
Affiliation(s)
- Ruirui Gui
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Wanqiao Li
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Zhipeng Li
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Hongbin Wang
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Yuchen Wu
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Wenlin Jiao
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Gang Zhao
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Yannan Shen
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Luping Wang
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Jialu Zhang
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Sihan Chen
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Linlin Hao
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China.
| | - Yunyun Cheng
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China.
| |
Collapse
|
37
|
Masliukov PM. Changes of Signaling Pathways in Hypothalamic Neurons with Aging. Curr Issues Mol Biol 2023; 45:8289-8308. [PMID: 37886966 PMCID: PMC10605528 DOI: 10.3390/cimb45100523] [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: 08/28/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
The hypothalamus is an important regulator of autonomic and endocrine functions also involved in aging regulation. The aging process in the hypothalamus is accompanied by disturbed intracellular signaling including insulin/insulin-like growth factor-1 (IGF-1)/growth hormone (GH), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT)/the mammalian target of rapamycin (mTOR), mitogen activated protein kinase (MAPK), janus kinase (JAK)/signal transducer and activator of transcription (STAT), AMP-activated protein kinase (AMPK), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ĸB), and nitric oxide (NO). In the current review, I have summarized the current understanding of the changes in the above-mentioned pathways in aging with a focus on hypothalamic alterations.
Collapse
Affiliation(s)
- Petr M Masliukov
- Department Normal Physiology, Yaroslavl State Medical University, ul. Revoliucionnaya 5, 150000 Yaroslavl, Russia
| |
Collapse
|