1
|
He T, Qin L, Chen S, Huo S, Li J, Zhang F, Yi W, Mei Y, Xiao G. Bone-derived factors mediate crosstalk between skeletal and extra-skeletal organs. Bone Res 2025; 13:49. [PMID: 40307216 PMCID: PMC12044029 DOI: 10.1038/s41413-025-00424-1] [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: 10/28/2024] [Revised: 03/15/2025] [Accepted: 03/20/2025] [Indexed: 05/02/2025] Open
Abstract
Bone has long been acknowledged as a fundamental structural entity that provides support and protection to the body's organs. However, emerging research indicates that bone plays a crucial role in the regulation of systemic metabolism. This is achieved through the secretion of a variety of hormones, cytokines, metal ions, extracellular vesicles, and other proteins/peptides, collectively referred to as bone-derived factors (BDFs). BDFs act as a medium through which bones can exert targeted regulatory functions upon various organs, thereby underscoring the profound and concrete implications of bone in human physiology. Nevertheless, there remains a pressing need for further investigations to elucidate the underlying mechanisms that inform the effects of bone on other body systems. This review aims to summarize the current findings related to the roles of these significant modulators across different organs and metabolic contexts by regulating critical genes and signaling pathways in vivo. It also addresses their involvement in the pathogenesis of various diseases affecting the musculoskeletal system, circulatory system, glucose and lipid metabolism, central nervous system, urinary system, and reproductive system. The insights gained from this review may contribute to the development of innovative therapeutic strategies through a focused approach to bone secretomes. Continued research into BDFs is expected to enhance our understanding of bone as a multifunctional organ with diverse regulatory roles in human health.
Collapse
Affiliation(s)
- Tailin He
- Department of Rheumatology and Immunology, Shenzhen Third People's Hospital, Shenzhen, 518112, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), 100101, Beijing, China
- Department of Biochemistry, Homeostatic Medicine Institute, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Lei Qin
- Department of Orthopedics, Shenzhen Nanshan People's Hospital, and the 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Sheng Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shaochuan Huo
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen 518000, China, Shenzhen Research Institute of Guangzhou University of Traditional Medicine (Futian), Shenzhen, 518000, China
| | - Jie Li
- Department of Biochemistry, Homeostatic Medicine Institute, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Fuping Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), 100101, Beijing, China
| | - Weihong Yi
- Department of Orthopedics, Shenzhen Nanshan People's Hospital, and the 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Yifang Mei
- Department of Rheumatology and Immunology, Shenzhen Third People's Hospital, Shenzhen, 518112, China.
| | - Guozhi Xiao
- Department of Biochemistry, Homeostatic Medicine Institute, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
2
|
Reichmann R, Nimptsch K, Pischon T, Gunter MJ, Jenab M, Eriksen AK, Tjonneland A, Janke J, Katzke V, Kaaks R, Schulze MB, Eichelmann F, Masala G, Sieri S, Pasanisi F, Tumino R, Giraudo MT, Rothwell J, Severi G, Jakszyn P, Sanchez-Perez MJ, Amiano P, Colorado-Yohar SM, Guevara M, van Guelpen B, Aglago EK, Heath AK, Smith-Byrne K, Weiderpass E, Aleksandrova K. Sex- and site-specific associations of circulating lipocalin 2 and incident colorectal cancer: Results from the EPIC cohort. Int J Cancer 2025; 156:930-942. [PMID: 39511728 DOI: 10.1002/ijc.35205] [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: 03/23/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 11/15/2024]
Abstract
Experimental research has uncovered lipocalin 2 (LCN2) as a novel biomarker implicated in the modulation of intestinal inflammation, metabolic homeostasis, and colon carcinogenesis. However, evidence from human research has been scant. We, therefore, explored the association of pre-diagnostic circulating LCN2 concentrations with incident colorectal cancer (CRC) in a nested case-control study within the in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. LCN2 was measured in 1267 incident CRC cases matched to 1267 controls using incidence density sampling. Conditional logistic regression was used to estimate incidence rate ratios (IRRs) and 95% confidence intervals (95% CIs) according to tumor subsite and sex. Weighted Cox proportional hazard regression was used to explore associations by adiposity status. In multivariable-adjusted analyses, the IRR [95% CI] per doubling in LCN2 concentration was 1.16 [0.98-1.37] for CRC overall, 1.26 [1.00-1.59] for colon cancer, and 1.08 [0.85-1.38] for rectal cancer. The association for colon cancer was more pronounced in women (IRR [95% CI], 1.66 [1.20-2.30]) and for proximal colon cancer (IRR [95% CI], 1.96 [1.15-3.34]), whereas no association was seen in men and distal colon cancer. The association for colon cancer was positive in individuals with high waist circumference (hazard ratio [95% CI], 1.69 [1.52-1.88]) and inverse in individuals with low waist circumference (hazard ratio [95% CI], 0.86 [0.76-0.98], P interaction<0.01). Overall, these data suggest that pre-diagnostic LCN2 concentrations were positively associated with colon cancer, particularly occurring in the proximal colon, in women and among individuals with abdominal adiposity.
Collapse
Grants
- Dutch Ministry of Public Health, Welfare and Sports (VWS), Netherlands Cancer Registry (NKR), LK Research Funds, Dutch Prevention Funds, Dutch ZON (Zorg Onderzoek Nederland), World Cancer Research Fund (WCRF), Statistics Netherlands (The Netherlands)
- German Cancer Aid, German Cancer Research Center (DKFZ), German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Federal Ministry of Education and Research (BMBF) (Germany)
- Danish Cancer Society (Denmark)
- Ligue Contre le Cancer, Institut Gustave Roussy, Mutuelle Générale de l'Education Nationale, Institut National de la Santé et de la Recherche Médicale (INSERM) (France)
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London
- 001 World Health Organization
- Associazione Italiana per la Ricerca sul Cancro-AIRC-Italy, Compagnia di SanPaolo and National Research Council (Italy)
- International Agency for Research on Cancer (IARC)
- Cancer Research UK (14,136 to EPIC-Norfolk; C8221/A29017 to EPIC-Oxford), Medical Research Council (1,000,143 to EPIC-Norfolk; MR/M012190/1 to EPIC-Oxford) (United Kingdom).
- Health Research Fund (FIS)-Instituto de Salud Carlos III (ISCIII), Regional Governments of Andalucía, Asturias, Basque Country, Murcia and Navarra, and the Catalan Institute of Oncology-ICO (Spain)
- Swedish Cancer Society, Swedish Research Council, Region Skåne and Region Västerbotten (Sweden)
Collapse
Affiliation(s)
- Robin Reichmann
- Biomarkers and Metabolism Research Group, Department of Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology, Bremen, Germany
| | - Katharina Nimptsch
- Molecular Epidemiology Research Group, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Biobank Technology Platform, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Tobias Pischon
- Molecular Epidemiology Research Group, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Biobank Technology Platform, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Core Facility Biobank, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marc J Gunter
- International Agency for Research on Cancer, World Health Organization, Lyon, France
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Mazda Jenab
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Anne Kirstine Eriksen
- Diet, Cancer and Health Research Group, Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark
| | - Anne Tjonneland
- Diet, Cancer and Health Research Group, Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Jürgen Janke
- Molecular Epidemiology Research Group, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Biobank Technology Platform, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Verena Katzke
- Department of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rudolf Kaaks
- Department of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Fabian Eichelmann
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Giovanna Masala
- Prevention and Clinical Network, Institute for the Study and Prevention of Cancer (ISPRO), Florenz, Italy
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Instituto Nazionale dei Tumori, Milan, Italy
| | - Fabrizio Pasanisi
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Rosario Tumino
- Hyblean Association for Epidemiological Research, Associazione Iblea per la Ricerca Epidemiologica (A.I.R.E.-ONLUS), Ragusa, Italy
| | - Maria Teresa Giraudo
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Joseph Rothwell
- CESP-Univ. Paris-Saclay, UVSQ, Inserm-"Exposome, heredity, cancer and health" Team, The Centre for Research in Epidemiology and Population Health, Villejuif, France
| | - Gianluca Severi
- CESP-Univ. Paris-Saclay, UVSQ, Inserm-"Exposome, heredity, cancer and health" Team, The Centre for Research in Epidemiology and Population Health, Villejuif, France
- Department of Statistics, Computer Science, Applications "G. Parenti" (DISIA), University of Florence, Florence, Italy
| | - Paula Jakszyn
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
- Blanquerna School of Health Sciences, Ramon Llull University, Barcelona, Spain
| | - Maria Jose Sanchez-Perez
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Public Health Research and Health Services Research Group, Andalusian School of Public Health (EASP), Granada, Andalucía, Spain
- Epidemiology, Prevention and Control of Cancer Research Group, Biosanitary Research Institute of Granada (ibs.Granada), Granada, Spain
- Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain
| | - Pilar Amiano
- Sub Directorate for Public Health and Addictions of Gipuzkoa, Ministry of Health of the Basque Government, San Sebastian, Spain
- Epidemiology of Chronic and Communicable Diseases Group, Biodonostia Health Research Institute, San Sebastian, Spain
- Instituto de Salud Carlos III, CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Sandra M Colorado-Yohar
- Department of Epidemiology, Murcia Regional Health Council, Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
- Research Group on Demography and Health, National Faculty of Public Health, University of Antioquia, Medellin, Colombia
| | - Marcela Guevara
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Epidemiology and Health Prevention Service, Institute of Public Health and Labor of Navarre, Pamplona, Navarra, Spain
- Epidemiology of Cancer and Other Chronic Diseases Research Group, Healthcare Research Institute of Navarre (IdiSNA), Pamplona, Spain
| | - Bethany van Guelpen
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Elom K Aglago
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Alicia K Heath
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Karl Smith-Byrne
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Krasimira Aleksandrova
- Biomarkers and Metabolism Research Group, Department of Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology, Bremen, Germany
- Faculty of Human and Health Sciences, University of Bremen, Bremen, Germany
| |
Collapse
|
3
|
Ernesto CMS, Laura SSD, Obed PMI, David AGR, Eloy GGJ, Lilia GHA. LCN2 blockade mitigating metabolic dysregulation and redefining appetite control in type 2 diabetes. Metab Brain Dis 2025; 40:97. [PMID: 39808380 PMCID: PMC11732943 DOI: 10.1007/s11011-024-01454-0] [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: 03/20/2024] [Accepted: 09/28/2024] [Indexed: 01/16/2025]
Abstract
LCN2 has an osteokine important for appetite regulation; in type 2 diabetes (T2D) it is not known whether appetite regulation mediated by LCN2 in the brain is altered. In this work, we focus on exploring the role of blocking LCN2 in metabolic health and appetite regulation within the central nervous system of mice with T2D. MATERIAL AND METHODS 4-week-old male C57BL/6 mice were used, divided into four experimental groups: intact, T2D, TD2/anti-LCN2, and T2D/IgG as isotype control. T2D was induced by low doses of streptozotocin and a high-carbohydrate diet. LCN2 blockade was performed by intraperitoneal administration of a polyclonal anti-LCN2 antibody. We analyzed metabolic parameters, food intake, feeding patterns, and serum LCN2 and leptin concentrations. In another group of intact or T2D mice, we analyzed the effect of blocking LCN2 and recombinant LCN2 on food consumption in a fasting-refeeding test and, the expression of cFOS and LCN2 in brain sections, specifically in the hypothalamus, piriform cortex, visceral area, arcuate nucleus and caudate-putamen. RESULTS T2D caused an increase in serum LCN2, without alterations in Ad libitum feeding, but with changes in the feeding pattern associated with alterations in LCN2-cFOS signalling in hypothalamic and non-hypothalamic brain regions. Blocking LCN2 improved metabolic parameters, increased Ad libitum feeding, and restored the feeding pattern after fasting, which is associated with enhanced LCN2 signalling in the brain. CONCLUSIONS Blocking LCN2 restores metabolic health and normalizes the pattern of food consumption by normalizing LCN2 signalling in different brain regions.
Collapse
Affiliation(s)
- Cifuentes-Mendiola Saúl Ernesto
- Section of Osteimmunology and Oral Immunology, Laboratory of Dental Reseach. FES Iztacala, National Autonomous University of Mexico (UNAM), México, Mexico State, México
| | - Sólis-Suarez Diana Laura
- Section of Osteimmunology and Oral Immunology, Laboratory of Dental Reseach. FES Iztacala, National Autonomous University of Mexico (UNAM), México, Mexico State, México
- Postgraduate in Dentistry Science, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Pérez-Martínez Isaac Obed
- Section of Sensation Neurobiology and Oral Movements, Laboratory of Dental Reseach. FES Iztacalaestigación Odontológica, National Autonomous University of Mexico (UNAM), México State, México, México
| | - Andrade-González Rey David
- Section of Sensation Neurobiology and Oral Movements, Laboratory of Dental Reseach. FES Iztacalaestigación Odontológica, National Autonomous University of Mexico (UNAM), México State, México, México
| | - García-Gama Jahaziel Eloy
- Section of Osteimmunology and Oral Immunology, Laboratory of Dental Reseach. FES Iztacala, National Autonomous University of Mexico (UNAM), México, Mexico State, México
| | - García-Hernández Ana Lilia
- Section of Osteimmunology and Oral Immunology, Laboratory of Dental Reseach. FES Iztacala, National Autonomous University of Mexico (UNAM), México, Mexico State, México.
| |
Collapse
|
4
|
Wat LW, Svensson KJ. Novel secreted regulators of glucose and lipid metabolism in the development of metabolic diseases. Diabetologia 2024; 67:2626-2636. [PMID: 39180580 DOI: 10.1007/s00125-024-06253-x] [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: 04/15/2024] [Accepted: 07/01/2024] [Indexed: 08/26/2024]
Abstract
The tight regulation of glucose and lipid metabolism is crucial for maintaining metabolic health. Dysregulation of these processes can lead to the development of metabolic diseases. Secreted factors, or hormones, play an essential role in the regulation of glucose and lipid metabolism, thus also playing an important role in the development of metabolic diseases such as type 2 diabetes and obesity. Given the important roles of secreted factors, there has been significant interest in identifying new secreted factors and new functions for existing secreted factors that control glucose and lipid metabolism. In this review, we evaluate novel secreted factors or novel functions of existing factors that regulate glucose and lipid metabolism discovered in the last decade, including secreted isoform of endoplasmic reticulum membrane complex subunit 10, vimentin, cartilage intermediate layer protein 2, isthmin-1, lipocalin-2, neuregulin-1 and neuregulin-4. We discuss their discovery, tissues of origin, mechanisms of action and sex differences, emphasising their potential to regulate metabolic processes central to diabetes. Additionally, we discuss the translational barriers, particularly the absence of identified receptors, that hamper their functional characterisation and further therapeutic development. Ultimately, the identification of new secreted factors may give insights into previously unidentified pathways of disease progression and mechanisms of glucose and lipid homeostasis.
Collapse
Affiliation(s)
- Lianna W Wat
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Katrin J Svensson
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
5
|
Sólis-Suarez DL, Cifuentes-Mendiola SE, González-Alva P, Rodríguez-Hernández AP, Martínez-Dávalos A, Llamosas-Hernandez FE, Godínez-Victoria M, García-Hernández AL. Lipocalin-2 as a fundamental protein in type 2 diabetes and periodontitis in mice. J Periodontol 2024. [PMID: 39189666 DOI: 10.1002/jper.24-0215] [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: 04/04/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Lipocalin-2 (LCN-2) is an osteokine that suppresses appetite, stimulates insulin secretion, regulates bone remodeling, and is induced by proinflammatory cytokines. The aim of this work was to investigate the participation of LCN-2 in periodontitis associated with type 2 diabetes (T2D) by evaluating alveolar bone loss, glycemic control, inflammation, and femur fragility. METHODS A murine model of periodontitis with T2D and elevated LCN-2 concentration was used. Functional LCN-2 inhibition was achieved using an anti-LCN-2 polyclonal antibody, and isotype immunoglobulin G was used as a control. The alveolar bone and femur were evaluated by micro-CT. Glucose metabolism was determined. Tumor necrosis factor (TNF-α) and receptor activator of nuclear factor kappa-B ligand (RANKL) levels in alveolar bone lysates were quantified using ELISA, and serum cytokines were quantified using flow cytometry. A three-point bending test was performed in the femur, and RANKL levels were measured in femur lysates using ELISA. RESULTS Functional inhibition of LCN-2 in T2D-periodontitis mice decreased alveolar bone loss in buccal and palatal surfaces and preserved the microarchitecture of the remaining bone, decreased TNF-α and RANKL in alveolar bone, reduced hyperglycemia, glucose intolerance, and insulin resistance, and increased insulin production through improving the functionality of pancreatic β cells. Furthermore, this inhibition increased serum free-glycerol levels, decreased serum interleukin (IL)-6, increased serum IL-4, and reduced femur fragility and RANKL expression in the femur. CONCLUSIONS LCN-2 participates in periodontitis associated with T2D. Inhibiting its function in mice with T2D and periodontitis improves pancreatic β-cell function, and glucose metabolism and decreases inflammatory cytokines and bone-RANKL levels, which results in the preservation of femoral and alveolar bone microarchitecture. PLAIN LANGUAGE SUMMARY In this study, we explored the role of a bone protein known as lipocalin-2 (LCN-2) in the connection between periodontitis and type 2 diabetes (T2D). Periodontitis is a destructive gum and alveolar bone disease. LCN-2 levels are increased in both T2D and periodontitis. Using a mouse model of T2D with periodontitis, we examined how blocking LCN-2 function affected various aspects of these two diseases. We found that this inhibition led to significant improvements. First, it reduced alveolar bone loss and preserved bone structure by decreasing local inflammation and bone resorption. Second, it improved glucose and lipid metabolism, leading to better blood-sugar control and decreased insulin resistance. Blocking the functions of LCN-2 also decreased systemic inflammation throughout the body and strengthened bone integrity. Overall, our results suggest that LCN-2 plays a crucial role in the periodontitis associated with T2D. By inhibiting LCN-2 function, we were able to improve pancreatic function, improve glucose metabolism, reduce inflammation, and enhance bone health. Targeting LCN-2 could be a promising strategy for the harmful effects of T2D and periodontitis.
Collapse
Affiliation(s)
- Diana Laura Sólis-Suarez
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico (UNAM), State of Mexico, Mexico, Mexico
- Postgraduate Course in Dental Sciences, National Autonomous University of Mexico, Mexico City, Mexico
| | - Saúl Ernesto Cifuentes-Mendiola
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico (UNAM), State of Mexico, Mexico, Mexico
| | - Patricia González-Alva
- Laboratory of Tissue Bioengineering, Faculty of Dentistry, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | | | - Arnulfo Martínez-Dávalos
- Endo-periodontology Department, Physics Institute, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | | | - Marycarmen Godínez-Victoria
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Ana Lilia García-Hernández
- Laboratory of Dental Research, Section of Osteoimmunology and Oral Immunology, FES Iztacala, National Autonomous University of Mexico (UNAM), State of Mexico, Mexico, Mexico
| |
Collapse
|
6
|
Xiao H, Li W, Qin Y, Lin Z, Qian C, Wu M, Xia Y, Bai J, Geng D. Crosstalk between Lipid Metabolism and Bone Homeostasis: Exploring Intricate Signaling Relationships. RESEARCH (WASHINGTON, D.C.) 2024; 7:0447. [PMID: 39165638 PMCID: PMC11334918 DOI: 10.34133/research.0447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024]
Abstract
Bone is a dynamic tissue reshaped by constant bone formation and bone resorption to maintain its function. The skeletal system accounts for approximately 70% of the total volume of the body, and continuous bone remodeling requires quantities of energy and material consumption. Adipose tissue is the main energy storehouse of the body and has a strong adaptive capacity to participate in the regulation of various physiological processes. Considering that obesity and metabolic syndrome have become major public health challenges, while osteoporosis and osteoporotic fractures have become other major health problems in the aging population, it would be interesting to explore these 2 diseases together. Currently, an increasing number of researchers are focusing on the interactions between multiple tissue systems, i.e., multiple organs and tissues that are functionally coordinated together and pathologically pathologically interact with each other in the body. However, there is lack of detailed reviews summarizing the effects of lipid metabolism on bone homeostasis and the interactions between adipose tissue and bone tissue. This review provides a detailed summary of recent advances in understanding how lipid molecules and adipose-derived hormones affect bone homeostasis, how bone tissue, as a metabolic organ, affects lipid metabolism, and how lipid metabolism is regulated by bone-derived cytokines.
Collapse
Affiliation(s)
- Haixiang Xiao
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei 230022, China
| | - Wenming Li
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yi Qin
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Zhixiang Lin
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Chen Qian
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Mingzhou Wu
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yu Xia
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jiaxiang Bai
- Department of Orthopedics, Jingjiang People’s Hospital Affiliated to Yangzhou University, Jingjiang 214500, Jiangsu Province, China
| | - Dechun Geng
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| |
Collapse
|
7
|
Afridi R, Kim JH, Bhusal A, Lee WH, Suk K. Lipocalin-2 as a mediator of neuroimmune communication. J Leukoc Biol 2024; 116:357-368. [PMID: 38149462 DOI: 10.1093/jleuko/qiad157] [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: 10/06/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023] Open
Abstract
Lipocalin-2, a neutrophil gelatinase-associated lipocalin, is a 25-kDa secreted protein implicated in a broad range of inflammatory diseases affecting the brain and periphery. It is a pleotropic protein expressed by various immune and nonimmune cells throughout the body. Importantly, the surge in lipocalin-2 levels in disease states has been associated with a myriad of undesirable effects, further exacerbating the ongoing pathological processes. In the brain, glial cells are the principal source of lipocalin-2, which plays a definitive role in determining their functional phenotypes. In different central nervous system pathologies, an increased expression of glial lipocalin-2 has been linked to neurotoxicity. Lipocalin-2 mediates a crosstalk between central and peripheral immune cells under neuroinflammatory conditions. One intriguing aspect is that elevated lipocalin-2 levels in peripheral disorders, such as cancer, metabolic conditions, and liver diseases, potentially incite an inflammatory activation of glial cells while disrupting neuronal functions. This review comprehensively summarizes the influence of lipocalin-2 on the exacerbation of neuroinflammation by regulating various cellular processes. Additionally, this review explores lipocalin-2 as a mediator of neuroimmune crosstalk in various central nervous system pathologies and highlights the role of lipocalin-2 in carrying inflammatory signals along the neuroimmune axis.
Collapse
Affiliation(s)
- Ruqayya Afridi
- Department of Pharmacology, School of Medicine, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41940, Republic of Korea
| | - Jae-Hong Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41940, Republic of Korea
| | - Anup Bhusal
- Department of Pharmacology, School of Medicine, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41940, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 plus KNU Creative BioResearch Group, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41944, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41940, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, 680 Gukchaebosang Street, Joong-gu, Daegu 41944, Republic of Korea
| |
Collapse
|
8
|
Yang Y, Liu J, Kousteni S. Lipocalin 2-A bone-derived anorexigenic and β-cell promoting signal: From mice to humans. J Diabetes 2024; 16:e13504. [PMID: 38035773 PMCID: PMC10940901 DOI: 10.1111/1753-0407.13504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/16/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
The skeleton is traditionally known for its structural support, organ protection, movement, and maintenance of mineral homeostasis. Over the last 10 years, bone has emerged as an endocrine organ with diverse physiological functions. The two key molecules in this context are fibroblast growth factor 23 (FGF23), secreted by osteocytes, and osteocalcin, a hormone produced by osteoblasts. FGF23 affects mineral homeostasis through its actions on the kidneys, and osteocalcin has beneficial effects in improving glucose homeostasis, muscle function, brain development, cognition, and male fertility. In addition, another osteoblast-derived hormone, lipocalin 2 (LCN2) has emerged into the researchers' field of vision. In this review, we mainly focus on LCN2's role in appetite regulation and glucose metabolism and also briefly introduce its effects in other pathophysiological conditions, such as nonalcoholic fatty liver disease, sarcopenic obesity, and cancer-induced cachexia.
Collapse
Affiliation(s)
- Yuying Yang
- Department of Endocrine and Metabolic Diseases, Rui‐jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai Institute of Endocrine and Metabolic Diseases, and Shanghai Clinical Center for Endocrine and Metabolic DiseasesShanghaiChina
- Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Clinical Research Center for Metabolic Diseases, Shanghai National Center for Translational Medicine, Rui‐jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jianmin Liu
- Department of Endocrine and Metabolic Diseases, Rui‐jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai Institute of Endocrine and Metabolic Diseases, and Shanghai Clinical Center for Endocrine and Metabolic DiseasesShanghaiChina
- Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Clinical Research Center for Metabolic Diseases, Shanghai National Center for Translational Medicine, Rui‐jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Stavroula Kousteni
- Department of Physiology and Cellular BiophysicsColumbia University Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
9
|
Zheng WB, Hu J, Sun L, Liu JY, Zhang Q, Wang O, Jiang Y, Xia WB, Xing XP, Li M. Correlation of lipocalin 2 and glycolipid metabolism and body composition in a large cohort of children with osteogenesis imperfecta. J Endocrinol Invest 2024; 47:47-58. [PMID: 37326909 PMCID: PMC10776749 DOI: 10.1007/s40618-023-02121-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/18/2023] [Indexed: 06/17/2023]
Abstract
PURPOSE Lipocalin 2 (LCN2) is a newly recognized bone-derived factor that is important in regulation of energy metabolism. We investigated the correlation of serum LCN2 levels and glycolipid metabolism, and body composition in a large cohort of patients with osteogenesis imperfecta (OI). METHODS A total of 204 children with OI and 66 age- and gender-matched healthy children were included. Circulating levels of LCN2 and osteocalcin were measured by enzyme-linked immunosorbent assay. Serum levels of fasting blood glucose (FBG), triglyceride (TG), total cholesterol (TC), and low- and high-density lipoprotein cholesterol (LDL-C, HDL-C) were measured by automated chemical analyzers. The body composition was measured by dual-energy X-ray absorptiometry. Grip strength and timed-up-and-go (TUG) were tested to evaluate the muscle function. RESULTS Serum LCN2 levels were 37.65 ± 23.48 ng/ml in OI children, which was significantly lower than those in healthy control (69.18 ± 35.43 ng/ml, P < 0.001). Body mass index (BMI) and serum FBG level were significantly higher and HDL-C levels were lower in OI children than healthy control (all P < 0.01). Grip strength was significantly lower (P < 0.05), and the TUG was significantly longer in OI patients than healthy control (P < 0.05). Serum LCN2 level was negatively correlated to BMI, FBG, HOMA-IR, HOMA-β, total body, and trunk fat mass percentage, and positively correlated to total body and appendicular lean mass percentage (all P < 0.05). CONCLUSIONS Insulin resistance, hyperglycemia, obesity, and muscle dysfunction are common in OI patients. As a novel osteogenic cytokine, LCN2 deficiency may be relevant to disorders of glucose and lipid metabolism, and dysfunction of muscle in OI patients.
Collapse
Affiliation(s)
- W-B Zheng
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100730, China
- Department of Endocrinology, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - J Hu
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100730, China
| | - L Sun
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100730, China
| | - J-Y Liu
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100730, China
| | - Q Zhang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100730, China
| | - O Wang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100730, China
| | - Y Jiang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100730, China
| | - W-B Xia
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100730, China
| | - X-P Xing
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100730, China
| | - M Li
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No. 1, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
10
|
Simpson CA, Santoro AM, Carpenter TO, Deng Y, Parziale S, Insogna KL. Circulating Levels of Leptin and Lipocalin-2 in Patients With X-Linked Hypophosphatemia. J Endocr Soc 2023; 7:bvad116. [PMID: 37860221 PMCID: PMC10583534 DOI: 10.1210/jendso/bvad116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Indexed: 10/21/2023] Open
Abstract
Individuals with X-linked hypophosphatemia (XLH) are at greater risk for being overweight or obese. Whether there are underlying metabolic abnormalities that put patients with XLH at greater risk for excessive weight gain is largely unknown. Lipocalin-2 (LCN2) has recently received attention as a factor regulating energy consumption and specifically is postulated to be anorexigenic and to improve insulin sensitivity. In a retrospective study, circulating levels of LCN2, leptin, and insulin were measured in 32 patients with XLH, ages 2-60 years, all of whom were being treated with burosumab, and 38 control subjects. Control subjects were chosen who were close in age to those with XLH, with a similar number of participants of each sex. Subjects were analyzed in 3 age cohorts, 2-10 years, 11-18 years, and 20-60 years. In all age groups LCN2 levels were lower in the patients with XLH than in controls but when adjusted for weight class (normal, overweight, obese) the differences were not significant. In contrast, serum leptin levels were significantly lower in children with XLH compared to controls in the 2-10 years age cohort. Serum levels of insulin were also significantly lower in the 2-10-year-old children with XLH when compared with controls. We conclude that changes in expression of lipocalin-2 in children and adolescents with XLH is unlikely to contribute to their risk for obesity in adulthood. It is unclear if lower circulating levels of leptin in these children plays a role in the higher prevalence of obesity among adults with XLH.
Collapse
Affiliation(s)
| | - Anna Maria Santoro
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| | | | - Yanhong Deng
- School of Public Health, Yale University, New Haven, CT 06510, USA
| | - Stephen Parziale
- School of Public Health, Yale University, New Haven, CT 06510, USA
| | - Karl L Insogna
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
11
|
Schröder SK, Gasterich N, Weiskirchen S, Weiskirchen R. Lipocalin 2 receptors: facts, fictions, and myths. Front Immunol 2023; 14:1229885. [PMID: 37638032 PMCID: PMC10451079 DOI: 10.3389/fimmu.2023.1229885] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 07/18/2023] [Indexed: 08/29/2023] Open
Abstract
The human 25-kDa Lipocalin 2 (LCN2) was first identified and purified as a protein that in part is associated with gelatinase from neutrophils. This protein shows a high degree of sequence similarity with the deduced sequences of rat α2-microglobulin-related protein and the mouse protein 24p3. Based on its typical lipocalin fold, which consists of an eight-stranded, anti-parallel, symmetrical β-barrel fold structure it was initially thought that LCN2 is a circulating protein functioning as a transporter of small lipophilic molecules. However, studies in Lcn2 null mice have shown that LCN2 has bacteriostatic properties and plays a key role in innate immunity by sequestering bacterial iron siderophores. Numerous reports have further shown that LCN2 is involved in the control of cell differentiation, energy expenditure, cell death, chemotaxis, cell migration, and many other biological processes. In addition, important roles for LCN2 in health and disease have been identified in Lcn2 null mice and multiple molecular pathways required for regulation of Lcn2 expression have been identified. Nevertheless, although six putative receptors for LCN2 have been proposed, there is a fundamental lack in understanding of how these cell-surface receptors transmit and amplify LCN2 to the cell. In the present review we summarize the current knowledge on LCN2 receptors and discuss inconsistencies, misinterpretations and false assumptions in the understanding of these potential LCN2 receptors.
Collapse
Affiliation(s)
- Sarah K. Schröder
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Natalie Gasterich
- Institute of Neuroanatomy, RWTH University Hospital Aachen, Aachen, Germany
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
12
|
Khosla S. Evidence in Humans for Bone as an Endocrine Organ Regulating Energy Metabolism. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2023; 31:100471. [PMID: 37576432 PMCID: PMC10417886 DOI: 10.1016/j.coemr.2023.100471] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
There is increasing evidence from animal models that bone, in addition to its traditional function of providing structural support for the organism, has a rich network of interactions with multiple other tissues. This perspective focuses on evidence from human studies demonstrating that bone is an endocrine organ regulating energy metabolism, with the specific examples being osteocalcin, lipocalin 2, RANKL, and sclerostin. Conversely, animal studies have also demonstrated that a key hormone regulating energy metabolism, leptin, regulates bone metabolism via the sympathetic nervous system. Studies in humans have established a role for the sympathetic nervous system in regulating bone turnover; indeed, the potential therapeutic benefit of targeting this pathway in humans to prevent postmenopausal bone loss is currently being evaluated.
Collapse
Affiliation(s)
- Sundeep Khosla
- Kogod Center on Aging and Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
13
|
Bini J. The historical progression of positron emission tomography research in neuroendocrinology. Front Neuroendocrinol 2023; 70:101081. [PMID: 37423505 PMCID: PMC10530506 DOI: 10.1016/j.yfrne.2023.101081] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
The rapid and continual development of a number of radiopharmaceuticals targeting different receptor, enzyme and small molecule systems has fostered Positron Emission Tomography (PET) imaging of endocrine system actions in vivo in the human brain for several decades. PET radioligands have been developed to measure changes that are regulated by hormone action (e.g., glucose metabolism, cerebral blood flow, dopamine receptors) and actions within endocrine organs or glands such as steroids (e.g., glucocorticoids receptors), hormones (e.g., estrogen, insulin), and enzymes (e.g., aromatase). This systematic review is targeted to the neuroendocrinology community that may be interested in learning about positron emission tomography (PET) imaging for use in their research. Covering neuroendocrine PET research over the past half century, researchers and clinicians will be able to answer the question of where future research may benefit from the strengths of PET imaging.
Collapse
Affiliation(s)
- Jason Bini
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States.
| |
Collapse
|
14
|
Deng M, Aberle MR, van Bijnen AAJHM, van der Kroft G, Lenaerts K, Neumann UP, Wiltberger G, Schaap FG, Olde Damink SWM, Rensen SS. Lipocalin-2 and neutrophil activation in pancreatic cancer cachexia. Front Immunol 2023; 14:1159411. [PMID: 37006254 PMCID: PMC10057111 DOI: 10.3389/fimmu.2023.1159411] [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: 02/05/2023] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundCancer cachexia is a multifactorial syndrome characterized by body weight loss and systemic inflammation. The characterization of the inflammatory response in patients with cachexia is still limited. Lipocalin-2, a protein abundant in neutrophils, has recently been implicated in appetite suppression in preclinical models of pancreatic cancer cachexia. We hypothesized that lipocalin-2 levels could be associated with neutrophil activation and nutritional status of pancreatic ductal adenocarcinoma (PDAC) patients.MethodsPlasma levels of neutrophil activation markers calprotectin, myeloperoxidase, elastase, and bactericidal/permeability-increasing protein (BPI) were compared between non-cachectic PDAC patients (n=13) and cachectic PDAC patients with high (≥26.9 ng/mL, n=34) or low (<26.9 ng/mL, n=34) circulating lipocalin-2 levels. Patients’ nutritional status was assessed by the patient-generated subjective global assessment (PG-SGA) and through body composition analysis using CT-scan slices at the L3 level.ResultsCirculating lipocalin-2 levels did not differ between cachectic and non-cachectic PDAC patients (median 26.7 (IQR 19.7-34.8) vs. 24.8 (16.6-29.4) ng/mL, p=0.141). Cachectic patients with high systemic lipocalin-2 levels had higher concentrations of calprotectin, myeloperoxidase, and elastase than non-cachectic patients or cachectic patients with low lipocalin-2 levels (calprotectin: 542.3 (355.8-724.9) vs. 457.5 (213.3-606.9), p=0.448 vs. 366.5 (294.5-478.5) ng/mL, p=0.009; myeloperoxidase: 30.3 (22.1-37.9) vs. 16.3 (12.0-27.5), p=0.021 vs. 20.2 (15.0-29.2) ng/mL, p=0.011; elastase: 137.1 (90.8-253.2) vs. 97.2 (28.8-215.7), p=0.410 vs. 95.0 (72.2-113.6) ng/mL, p=0.006; respectively). The CRP/albumin ratio was also higher in cachectic patients with high lipocalin-2 levels (2.3 (1.3-6.0) as compared to non-cachectic patients (1.0 (0.7-4.2), p=0.041). Lipocalin-2 concentrations correlated with those of calprotectin (rs=0.36, p<0.001), myeloperoxidase (rs=0.48, p<0.001), elastase (rs=0.50, p<0.001), and BPI (rs=0.22, p=0.048). Whereas no significant correlations with weight loss, BMI, or L3 skeletal muscle index were observed, lipocalin-2 concentrations were associated with subcutaneous adipose tissue index (rs=-0.25, p=0.034). Moreover, lipocalin-2 tended to be elevated in severely malnourished patients compared with well-nourished patients (27.2 (20.3-37.2) vs. 19.9 (13.4-26.4) ng/mL, p=0.058).ConclusionsThese data suggest that lipocalin-2 levels are associated with neutrophil activation in patients with pancreatic cancer cachexia and that it may contribute to their poor nutritional status.
Collapse
Affiliation(s)
- Min Deng
- Department of Surgery and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
| | - Merel R. Aberle
- Department of Surgery and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
| | - Annemarie A. J. H. M. van Bijnen
- Department of Surgery and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
| | - Gregory van der Kroft
- Department of Surgery and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
- Department of General, Visceral- and Transplantation Surgery, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Kaatje Lenaerts
- Department of Surgery and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
| | - Ulf P. Neumann
- Department of Surgery and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
- Department of General, Visceral- and Transplantation Surgery, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Georg Wiltberger
- Department of General, Visceral- and Transplantation Surgery, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Frank G. Schaap
- Department of Surgery and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
- Department of General, Visceral- and Transplantation Surgery, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Steven W. M. Olde Damink
- Department of Surgery and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
- Department of General, Visceral- and Transplantation Surgery, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Sander S. Rensen
- Department of Surgery and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
- *Correspondence: Sander S. Rensen,
| |
Collapse
|
15
|
Dai L, Liu Z, Guo L, Chai Y, Yang Y, Wang Y, Ma Y, Shi C, Zhang W. Multi-Tissue Transcriptome Study of Innate Immune Gene Expression Profiling Reveals Negative Energy Balance Altered the Defense and Promoted System Inflammation of Dairy Cows. Vet Sci 2023; 10:vetsci10020107. [PMID: 36851411 PMCID: PMC9959304 DOI: 10.3390/vetsci10020107] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023] Open
Abstract
Negative energy balance (NEB) during the perinatal period leads to metabolic and immunological disorders in dairy cows, resulting in systemic responses and inflammation. The innate immune system is crucial for the host's protection and inflammatory response. However, systematic research is still lacking on how NEB affects the innate immune system to alter the 'host defense capability and inflammatory response. In this investigation, raw transcriptome data of adipose, blood, endometrial, hypothalamus, and liver tissues were downloaded from a public database, cleaned, aligned, quantified, and batch-corrected. The innate immune gene list was retrieved from innateDB, followed by the expression matrix of innate immune genes in various tissues for differential expression analysis, principle component analysis (PCA), and gene set enrichment analysis (GSEA). Under the effect of NEB, adipose tissue had the most differentially expressed genes, which were predominantly up-regulated, whereas blood GSEA had the most enriched biological processes, which were predominantly down-regulated. The gene sets shared by different tissues, which are predominantly involved in biological processes associated with defense responses and inflammation, were dramatically down-regulated in endometrial tissues and highly up-regulated in other tissues. Under the impact of NEB, LBP, PTX3, S100A12, and LCN2 play essential roles in metabolism and immunological control. In conclusion, NEB can downregulate the defensive response of innate immune genes in endometrial, upregulate the immune and inflammatory response of other tissues, activate the host defense response, and increase the systemic inflammatory response. The analysis of the effects of NEB on innate immune genes from the multiple tissues analysis provides new insights into the crosstalk between metabolism and immunity and also provides potential molecular targets for disease diagnosis and disease resistance breeding in dairy cows.
Collapse
Affiliation(s)
- Lingli Dai
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
- Veterinary Research Institute, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot 010031, China
| | - Zaixia Liu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lili Guo
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yuan Chai
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yanda Yang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yu Wang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yanfen Ma
- Veterinary Research Institute, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot 010031, China
- School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Caixia Shi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
- Correspondence: (C.S.); (W.Z.)
| | - Wenguang Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Engineering Research Center of Genomic Big Data for Agriculture, Hohhot 010018, China
- College of Life Science, Inner Mongolia Agricultural University, Hohhot 010018, China
- Correspondence: (C.S.); (W.Z.)
| |
Collapse
|
16
|
Roberts JL, Golloshi M, Harding DB, Conduah M, Liu G, Drissi H. Bifidobacterium longum supplementation improves age-related delays in fracture repair. Aging Cell 2023; 22:e13786. [PMID: 36704918 PMCID: PMC10086533 DOI: 10.1111/acel.13786] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 01/28/2023] Open
Abstract
Age-related delays in bone repair remains an important clinical issue that can prolong pain and suffering. It is now well established that inflammation increases with aging and that this exacerbated inflammatory response can influence skeletal regeneration. Recently, simple dietary supplementation with beneficial probiotic bacteria has been shown to influence fracture repair in young mice. However, the contribution of the gut microbiota to age-related impairments in fracture healing remains unknown. Here, we sought to determine whether supplementation with a single beneficial probiotic species, Bifidobacterium longum (B. longum), would promote fracture repair in aged (18-month-old) female mice. We found that B. longum supplementation accelerated bony callus formation which improved mechanical properties of the fractured limb. We attribute these pro-regenerative effects of B. longum to preservation of intestinal barrier, dampened systemic inflammation, and maintenance of the microbiota community structure. Moreover, B. longum attenuated many of the fracture-induced systemic pathologies. Our study provides evidence that targeting the gut microbiota using simple dietary approaches can improve fracture healing outcomes and minimize systemic pathologies in the context of aging.
Collapse
Affiliation(s)
- Joseph L Roberts
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta VA Health Care System, Decatur, Georgia, USA
| | - Mateo Golloshi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta VA Health Care System, Decatur, Georgia, USA
| | - Derek B Harding
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta VA Health Care System, Decatur, Georgia, USA
| | - Madison Conduah
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta VA Health Care System, Decatur, Georgia, USA
| | - Guanglu Liu
- Atlanta VA Health Care System, Decatur, Georgia, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA.,Atlanta VA Health Care System, Decatur, Georgia, USA
| |
Collapse
|
17
|
Sahu B, Bal NC. Adipokines from white adipose tissue in regulation of whole body energy homeostasis. Biochimie 2023; 204:92-107. [PMID: 36084909 DOI: 10.1016/j.biochi.2022.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/08/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023]
Abstract
Diseases originating from altered energy homeostasis including obesity, and type 2 diabetes are rapidly increasing worldwide. Research in the last few decades on animal models and humans demonstrates that the white adipose tissue (WAT) is critical for energy balance and more than just an energy storage site. WAT orchestrates the whole-body metabolism through inter-organ crosstalk primarily mediated by cytokines named "Adipokines". The adipokines influence metabolism and fuel selection of the skeletal muscle and liver thereby fine-tuning the load on WAT itself in physiological conditions like starvation, exercise and cold. In addition, adipokine secretion is influenced by various pathological conditions like obesity, inflammation and diabetes. In this review, we have surveyed the current state of knowledge on important adipokines and their significance in regulating energy balance and metabolic diseases. Furthermore, we have summarized the interplay of pro-inflammatory and anti-inflammatory adipokines in the modulation of pathological conditions.
Collapse
Affiliation(s)
- Bijayashree Sahu
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India.
| | - Naresh C Bal
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
18
|
Yuan XC, Tao YX. Ligands for Melanocortin Receptors: Beyond Melanocyte-Stimulating Hormones and Adrenocorticotropin. Biomolecules 2022; 12:biom12101407. [PMID: 36291616 PMCID: PMC9599618 DOI: 10.3390/biom12101407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
The discovery of melanocortins in 1916 has resulted in more than 100 years of research focused on these peptides. Extensive studies have elucidated well-established functions of melanocortins mediated by cell surface receptors, including MSHR (melanocyte-stimulating hormone receptor) and ACTHR (adrenocorticotropin receptor). Subsequently, three additional melanocortin receptors (MCRs) were identified. Among these five MCRs, MC3R and MC4R are expressed primarily in the central nervous system, and are therefore referred to as the neural MCRs. Since the central melanocortin system plays important roles in regulating energy homeostasis, targeting neural MCRs is emerging as a therapeutic approach for treating metabolic conditions such as obesity and cachexia. Early efforts modifying endogenous ligands resulted in the development of many potent and selective ligands. This review focuses on the ligands for neural MCRs, including classical ligands (MSH and agouti-related peptide), nonclassical ligands (lipocalin 2, β-defensin, small molecules, and pharmacoperones), and clinically approved ligands (ACTH, setmelanotide, bremelanotide, and several repurposed drugs).
Collapse
Affiliation(s)
- Xiao-Chen Yuan
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230061, China
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
- Correspondence:
| |
Collapse
|
19
|
Parker L, Ang T, Morrison DJ, Lee NJ, Levinger I, Keske MA. Prior aerobic exercise mitigates the decrease in serum osteoglycin and lipocalin-2 following high-glucose mixed-nutrient meal ingestion in young men. Am J Physiol Endocrinol Metab 2022; 323:E319-E332. [PMID: 35767699 DOI: 10.1152/ajpendo.00025.2022] [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] [Indexed: 11/22/2022]
Abstract
Osteoglycin (OGN) and lipocalin-2 (LCN2) are hormones that can be secreted by bone and have been linked to glucose homeostasis in rodents. However, the endocrine role of these hormones in humans is contradictory and unclear. We examined the effects of exercise and meal ingestion on circulating serum OGN and LCN2 levels in eight healthy males {age: 28 [25, 30] years [median ± interquartile range (IQR)] and body mass index [BMI]: 24.3 [23.6, 25.5] kg/m2}. In a randomized crossover design, participants ingested a high-glucose (1.1 g glucose/kg body wt) mixed-nutrient meal (45% carbohydrate, 20% protein, and 35% fat) on a rest-control day and 3 and 24 h after aerobic cycling exercise (1 h at 70%-75% V̇o2peak). Acute aerobic exercise increased serum LCN2 levels immediately after exercise (∼61%), which remained elevated 3-h postexercise (∼55%). In contrast, serum OGN remained similar to baseline levels throughout the 3-h postexercise recovery period. The ingestion of a high-glucose mixed-nutrient meal led to a decrease in serum OGN at 90-min (approximately -17%) and 120-min postprandial (approximately -44%), and a decrease in LCN2 at 120-min postprandial (approximately -26%). Compared with the control meal, prior exercise elevated serum OGN and LCN2 levels at 120-min postprandial when the meal was ingested 3-h (OGN: ∼74% and LCN2: ∼68%) and 24-h postexercise (OGN: ∼56% and LCN2: ∼16%). Acute exercise increases serum LCN2 and attenuates the postprandial decrease in OGN and LCN2 following high-glucose mixed-nutrient meal ingestion. The potential endocrine role of circulating OGN and LCN2 in humans warrants further investigation.NEW & NOTEWORTHY We provide novel evidence that OGN and LCN2 decrease 120 min after ingesting a high-glucose mixed-nutrient meal in healthy adults. Acute aerobic exercise increases circulating LCN2 for up to 3-h postexercise, whereas circulating OGN remains similar to baseline. Despite differing postexercise responses, postprandial LCN2 and OGN are elevated when the high-glucose meal is ingested 3-h and 24-h postexercise. Findings support that OGN and LCN2 are dynamically linked to energy homeostasis in humans.
Collapse
Affiliation(s)
- Lewan Parker
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Teddy Ang
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Dale J Morrison
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Nicola J Lee
- Garvan Institute of Medical Research, St. Vincent's Hospital, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Itamar Levinger
- Institute for Health and Sport (IHES), Victoria University, Footscray, Victoria, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, St Albans, Victoria, Australia
| | - Michelle A Keske
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
20
|
Wu X, Dai Y, Nie K. Research Progress of Liujunzi Decoction in the Treatment of Tumor-Associated Anorexia. Drug Des Devel Ther 2022; 16:1731-1741. [PMID: 35698654 PMCID: PMC9188393 DOI: 10.2147/dddt.s365292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/26/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Xipei Wu
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Yongzhao Dai
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Ke Nie
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
- Correspondence: Ke Nie, School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, People’s Republic of China, Email ;
| |
Collapse
|
21
|
Liu X, Zhang J, Yuan J, Ding R, Liu T, Jia J. LCN2 is a new diagnostic biomarker and potential therapeutic target in idiopathic short stature. J Cell Mol Med 2022; 26:3568-3581. [PMID: 35610759 PMCID: PMC9189333 DOI: 10.1111/jcmm.17408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 03/05/2022] [Accepted: 05/05/2022] [Indexed: 12/21/2022] Open
Abstract
Idiopathic short stature (ISS) is the most common paediatric endocrine disease. However, the underlying pathology of ISS remains unclear. Currently, there are no effective diagnostic markers or therapeutic strategies available for ISS. In this study, we aimed to identify differential plasma protein expression and novel biomarkers in patients with ISS, and elucidate the biological functions of candidate proteins in ISS pathogenesis. Four specimen pairs from four ISS children and age‐/sex‐matched control individuals were subjected to proteomics analysis, and 340 samples of children with a mean age 9.73 ± 0.24 years were utilized to further verify the differentially expressed proteins by enzyme‐linked immunosorbent assay (ELISA). The receiver‐operating characteristic (ROC) curve and the area under the ROC curve (AUC) were plotted. A total of 2040 proteins were identified, of which 84 were differentially expressed. In vitro and in vivo experiments confirmed the biological functions of these candidate proteins. LCN2 overexpression in ISS was verified using ELISA. Meanwhile, LCN2 showed high sensitivity and specificity in discriminating children with ISS from those with growth hormone deficiency, precocious puberty and normal control individuals. The upregulated expression of LCN2 not only suppressed food intake but also impaired chondrocyte proliferation and bone growth in chondrocytes and rats. As a result, the rats presented a short‐stature phenotype. Subsequently, we found that bone growth inhibition recovered after LCN2 overexpression was stopped in immature rats. To our knowledge, this is the first study to report that LCN2 may be a significant target for ISS diagnosis and treatment.
Collapse
Affiliation(s)
- Xijuan Liu
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jinghong Yuan
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rui Ding
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tao Liu
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jingyu Jia
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
22
|
Huisman C, Norgard MA, Levasseur PR, Krasnow SM, van der Wijst MGP, Olson B, Marks DL. Critical changes in hypothalamic gene networks in response to pancreatic cancer as found by single-cell RNA sequencing. Mol Metab 2022; 58:101441. [PMID: 35031523 PMCID: PMC8851272 DOI: 10.1016/j.molmet.2022.101441] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Cancer cachexia is a devastating chronic condition characterized by involuntary weight loss, muscle wasting, abnormal fat metabolism, anorexia, and fatigue. However, the molecular mechanisms underlying this syndrome remain poorly understood. In particular, the hypothalamus may play a central role in cachexia, given that it has direct access to peripheral signals because of its anatomical location and attenuated blood-brain barrier. Furthermore, this region has a critical role in regulating appetite and metabolism. METHODS To provide a detailed analysis of the hypothalamic response to cachexia, we performed single-cell RNA-seq combined with RNA-seq of the medial basal hypothalamus (MBH) in a mouse model for pancreatic cancer. RESULTS We found many cell type-specific changes, such as inflamed endothelial cells, stressed oligodendrocyes and both inflammatory and moderating microglia. Lcn2, a newly discovered hunger suppressing hormone, was the highest induced gene. Interestingly, cerebral treatment with LCN2 not only induced many of the observed molecular changes in cachexia but also affected gene expression in food-intake decreasing POMC neurons. In addition, we found that many of the cachexia-induced molecular changes found in the hypothalamus mimic those at the primary tumor site. CONCLUSION Our data reveal that multiple cell types in the MBH are affected by tumor-derived factors or host factors that are induced by tumor growth, leading to a marked change in the microenvironment of neurons critical for behavioral, metabolic, and neuroendocrine outputs dysregulated during cachexia. The mechanistic insights provided in this study explain many of the clinical features of cachexia and will be useful for future therapeutic development.
Collapse
Affiliation(s)
- Christian Huisman
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, United States; Knight Cancer Institute, Oregon Health & Science University, Portland, United States.
| | - Mason A Norgard
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, United States
| | - Peter R Levasseur
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, United States
| | - Stephanie M Krasnow
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, United States
| | - Monique G P van der Wijst
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Brennan Olson
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, United States; Medical Scientist Training Program, Oregon Health & Science University, Portland, United States
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, United States; Knight Cancer Institute, Oregon Health & Science University, Portland, United States; Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, United States.
| |
Collapse
|
23
|
Daoud MS, Hussain SD, Al-Daghri NM. Cardiometabolic associations of circulating Lipocalin-2 in adults with varying degrees of adiposity and insulin resistance. Arch Biochem Biophys 2022; 717:109138. [DOI: 10.1016/j.abb.2022.109138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 11/02/2022]
|
24
|
Li N, Xu B, Zeng J, Lei S, Gu L, Feng L, Zhu B, Huang Y, Wang L, Su L, Qu S, Cheng X, Bu L. Development of a New Index Based on Preoperative Serum Lipocalin 2 to Predict Post-LSG Weight Reduction. Obes Surg 2022; 32:1184-1192. [PMID: 35138515 PMCID: PMC8933383 DOI: 10.1007/s11695-022-05916-1] [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: 11/16/2021] [Revised: 01/03/2022] [Accepted: 01/14/2022] [Indexed: 11/30/2022]
Abstract
Background Bariatric surgery is the most effective therapy for obesity, but targeted weight reduction is not always achieved. Serum lipocalin-2 (LCN2) is closely associated with obesity, but its impact on weight loss after surgery is unknown. We aimed to access the reliability of LCN2 levels and other parameters as effective predictors of excellent weight loss (≥ 75% excess weight loss (EWL)) 1 year after bariatric surgery. Methods This retrospective study evaluated 450 patients (aged 18–65 years) with obesity at 3 months and 1 year after laparoscopic sleeve gastrectomy (LSG) surgery. Seventy-four patients who underwent LSG surgery and met the inclusion and exclusion criteria were included in this study. Serum LCN2, thyroid function, and metabolic and anthropometric parameters were assessed. Weight reduction was expressed as %EWL and percent total weight loss (%TWL) at 3 months and 1 year post surgery. Multivariable logistic regression analysis and receiver operating characteristic (ROC) curve analysis were used to evaluate predictors of ≥ 75%EWL. Results In our cohort, %EWL and %TWL were both strongly associated with preoperative serum LCN2 levels. The binary logistic regression analysis showed that preoperative LCN2, waist circumference, and glycated hemoglobin were independent predictors of excellent weight loss. Conclusions Based on these results, we determined a new P index with better predictive value for excellent weight reduction (≥ 75%EWL) 1 year after LSG surgery. Graphical abstract ![]()
Collapse
Affiliation(s)
- Nannan Li
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Bei Xu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Jiangping Zeng
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Shihui Lei
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Lei Gu
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Lijin Feng
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Bing Zhu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Yueye Huang
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Lu Wang
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Lili Su
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Xiaoyun Cheng
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, China.
| | - Le Bu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, China.
| |
Collapse
|
25
|
Ortuno MJ, Schneeberger M, Ilanges A, Marchildon F, Pellegrino K, Friedman JM, Ducy P. Melanocortin 4 receptor stimulation prevents anti-depressant-associated weight gain in mice caused by long-term fluoxetine exposure. J Clin Invest 2021; 131:151976. [PMID: 34673574 DOI: 10.1172/jci151976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/19/2021] [Indexed: 11/17/2022] Open
Abstract
Contrasting with the predicted anorexigenic effect of increasing brain serotonin signaling, long-term use of selective serotonin reuptake inhibitors (SSRIs) antidepressants correlates with body weight gain. This adverse outcome increases the risk of transitioning to obesity and interferes with treatment compliance. Here we show that orally administered fluoxetine (Flx), a widely prescribed SSRI, increased body weight by enhancing food intake in healthy mice at two different time points and through two distinct mechanisms. Within hours, Flx decreased the activity of a subset of brainstem serotonergic neurons by triggering autoinhibitory signaling through the Htr1a receptor. Upon longer treatment Flx blunted Htr2c expression/signaling, decreased the phosphorylation of Creb and Stat3 and dampened the production of POMC/α-MSH in hypothalamic neurons, thereby increasing food intake. Accordingly, exogenous stimulation of the melanocortin 4 receptor (MC4R) by co-treating mice with Flx and lipocalin-2, an anorexigenic hormone signaling through this receptor, normalized feeding and body weight. Flx and other SSRIs also inhibit CREB/STAT3 phosphorylation in a human neuronal cell line suggesting that these non-canonical effects could also occur in long-term users of SSRIs. By defining the molecular basis of the long-term SSRIs-associated weight gain this study proposes a therapeutic strategy to counter it.
Collapse
Affiliation(s)
- Maria Jose Ortuno
- Department of Genetics and Development, Columbia University, New York, United States of America
| | - Marc Schneeberger
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States of America
| | - Anoj Ilanges
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States of America
| | - François Marchildon
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, United States of America
| | - Kyle Pellegrino
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States of America
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States of America
| | - Patricia Ducy
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, United States of America
| |
Collapse
|
26
|
Adipose-Derived Lipid-Binding Proteins: The Good, the Bad and the Metabolic Diseases. Int J Mol Sci 2021; 22:ijms221910460. [PMID: 34638803 PMCID: PMC8508731 DOI: 10.3390/ijms221910460] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
Adipose tissue releases a large range of bioactive factors called adipokines, many of which are involved in inflammation, glucose homeostasis and lipid metabolism. Under pathological conditions such as obesity, most of the adipokines are upregulated and considered as deleterious, due to their pro-inflammatory, pro-atherosclerotic or pro-diabetic properties, while only a few are downregulated and would be designated as beneficial adipokines, thanks to their counteracting properties against the onset of comorbidities. This review focuses on six adipose-derived lipid-binding proteins that have emerged as key factors in the development of obesity and diabetes: Retinol binding protein 4 (RBP4), Fatty acid binding protein 4 (FABP4), Apolipoprotein D (APOD), Lipocalin-2 (LCN2), Lipocalin-14 (LCN14) and Apolipoprotein M (APOM). These proteins share structural homology and capacity to bind small hydrophobic molecules but display opposite effects on glucose and lipid metabolism. RBP4 and FABP4 are positively associated with metabolic syndrome, while APOD and LCN2 are ubiquitously expressed proteins with deleterious or beneficial effects, depending on their anatomical site of expression. LCN14 and APOM have been recently identified as adipokines associated with healthy metabolism. Recent findings on these lipid-binding proteins exhibiting detrimental or protective roles in human and murine metabolism and their involvement in metabolic diseases are also discussed.
Collapse
|
27
|
Dekens DW, Eisel ULM, Gouweleeuw L, Schoemaker RG, De Deyn PP, Naudé PJW. Lipocalin 2 as a link between ageing, risk factor conditions and age-related brain diseases. Ageing Res Rev 2021; 70:101414. [PMID: 34325073 DOI: 10.1016/j.arr.2021.101414] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022]
Abstract
Chronic (neuro)inflammation plays an important role in many age-related central nervous system (CNS) diseases, including Alzheimer's disease, Parkinson's disease and vascular dementia. Inflammation also characterizes many conditions that form a risk factor for these CNS disorders, such as physical inactivity, obesity and cardiovascular disease. Lipocalin 2 (Lcn2) is an inflammatory protein shown to be involved in different age-related CNS diseases, as well as risk factor conditions thereof. Lcn2 expression is increased in the periphery and the brain in different age-related CNS diseases and also their risk factor conditions. Experimental studies indicate that Lcn2 contributes to various neuropathophysiological processes of age-related CNS diseases, including exacerbated neuroinflammation, cell death and iron dysregulation, which may negatively impact cognitive function. We hypothesize that increased Lcn2 levels as a result of age-related risk factor conditions may sensitize the brain and increase the risk to develop age-related CNS diseases. In this review we first provide a comprehensive overview of the known functions of Lcn2, and its effects in the CNS. Subsequently, this review explores Lcn2 as a potential (neuro)inflammatory link between different risk factor conditions and the development of age-related CNS disorders. Altogether, evidence convincingly indicates Lcn2 as a key constituent in ageing and age-related brain diseases.
Collapse
Affiliation(s)
- Doortje W Dekens
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Leonie Gouweleeuw
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Regien G Schoemaker
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Peter P De Deyn
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Laboratory of Neurochemistry and Behaviour, Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Petrus J W Naudé
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands; Department of Psychiatry and Mental Health and Neuroscience Institute, Brain Behaviour Unit, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
28
|
Watanabe LM, Hashimoto AC, Torres DJ, Alfulaij N, Peres R, Sultana R, Maunakea AK, Berry MJ, Seale LA. Effect of statin treatment in obese selenium-supplemented mice lacking selenocysteine lyase. Mol Cell Endocrinol 2021; 533:111335. [PMID: 34052303 PMCID: PMC8263501 DOI: 10.1016/j.mce.2021.111335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/14/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022]
Abstract
People with obesity are often dyslipidemic and prescribed statins to prevent cardiovascular events. A common side effect of statin use is myopathy. This could potentially be caused by the reduction of selenoproteins that curb oxidative stress, in turn, affecting creatine metabolism. We determined if statins regulate hepatic and muscular selenoprotein expression, oxidative stress and creatine metabolism. Mice lacking selenocysteine lyase (Scly KO), a selenium-provider enzyme for selenoprotein synthesis, were fed a high-fat, Se-supplemented diet and treated with simvastatin. Statin improved creatine metabolism in females and oxidative responses in both sexes. Male Scly KO mice were heavier than females after statin treatment. Hepatic selenoproteins were unaffected by statin and genotype in females. Statin upregulated muscular Gpx1 in females but not males, while Scly loss downregulated muscular Gpx1 in males and Selenon in females. Osgin1 was reduced in statin-treated Scly KO males after AmpliSeq analysis. These results refine our understanding of the sex-dependent role of selenium in statin responses.
Collapse
Affiliation(s)
- Ligia M Watanabe
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96813, USA; Department of Internal Medicine, Faculty of Medicine of Ribeirão Preto, University of São Paulo - FMRP/USP, Brazil
| | - Ann C Hashimoto
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96813, USA
| | - Daniel J Torres
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96813, USA; Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Naghum Alfulaij
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96813, USA; Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Rafael Peres
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96813, USA
| | - Razvan Sultana
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96813, USA
| | - Alika K Maunakea
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96813, USA
| | - Marla J Berry
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Lucia A Seale
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
29
|
Naudé PJW, Ramakers IHGB, van der Flier WM, Jiskoot LC, Reesink FE, Claassen JAHR, Koek HL, Eisel ULM, De Deyn PP. Serum and cerebrospinal fluid Neutrophil gelatinase-associated lipocalin (NGAL) levels as biomarkers for the conversion from mild cognitive impairment to Alzheimer's disease dementia. Neurobiol Aging 2021; 107:1-10. [PMID: 34365256 DOI: 10.1016/j.neurobiolaging.2021.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/29/2021] [Accepted: 07/02/2021] [Indexed: 10/20/2022]
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL) is an acute phase protein that has been reported as a potential marker for pre-dementia stages of Alzheimer's disease (AD). Longitudinal studies for its association with the conversion of mild cognitive impairment to AD is still lacking. This study included n = 268 study participants with subjective cognitive decline (SCD) (n=82), mild cognitive impairment (MCI) (n=98) and AD dementia (n=88) at baseline and two-year follow-up clinical assessments. Serum and cerebrospinal fluid (CSF)NGAL, CSF amyloid beta1-42, total-Tau, and phospho-Tau levels were measured with ELISA analysis. CSF NGAL levels were significantly lower in MCI participants compared to people with SCD at baseline. Lower baseline CSF NGAL levels predicted MCI converters to AD dementia vs. non-converters after 2-years follow-up. A positive correlation between CSF NGAL and amyloid beta1-42 was found particularly in MCI participants at baseline. NGAL in CSF holds potential to be used as a predictive marker for the conversion of MCI to AD dementia and may reflect pathophysiological processes of prodromal AD neuropathology.
Collapse
Affiliation(s)
- Petrus J W Naudé
- Department of Neurology and Alzheimer Research Centre, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands; Department of Psychiatry and Mental Health and Neuroscience Institute, Brain Behavior Unit, University of Cape Town, Cape Town, South Africa.
| | - Inez H G B Ramakers
- Alzheimer Center Limburg, School for Mental Health and Neuroscience (MHeNS), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Wiesje M van der Flier
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Department of Epidemiology and Data Sciences, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Lize C Jiskoot
- Department of Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands; Dementia Research Centre, University College London, London, UK
| | - Fransje E Reesink
- Department of Neurology and Alzheimer Research Centre, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Jurgen A H R Claassen
- Departments of Neurology and Geriatrics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Huiberdina L Koek
- Radboud University Medical Center and Radboud Alzheimer Center, Department of Geriatrics, Nijmegen, The Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands; University Center of Psychiatry & Interdisciplinary Center of Psychopathology of Emotion Regulation, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peter P De Deyn
- Department of Neurology and Alzheimer Research Centre, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands; Laboratory of Neurochemistry and Behavior, Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
30
|
Stucker S, De Angelis J, Kusumbe AP. Heterogeneity and Dynamics of Vasculature in the Endocrine System During Aging and Disease. Front Physiol 2021; 12:624928. [PMID: 33767633 PMCID: PMC7987104 DOI: 10.3389/fphys.2021.624928] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
The endocrine system consists of several highly vascularized glands that produce and secrete hormones to maintain body homeostasis and regulate a range of bodily functions and processes, including growth, metabolism and development. The dense and highly vascularized capillary network functions as the main transport system for hormones and regulatory factors to enable efficient endocrine function. The specialized capillary types provide the microenvironments to support stem and progenitor cells, by regulating their survival, maintenance and differentiation. Moreover, the vasculature interacts with endocrine cells supporting their endocrine function. However, the structure and niche function of vasculature in endocrine tissues remain poorly understood. Aging and endocrine disorders are associated with vascular perturbations. Understanding the cellular and molecular cues driving the disease, and age-related vascular perturbations hold potential to manage or even treat endocrine disorders and comorbidities associated with aging. This review aims to describe the structure and niche functions of the vasculature in various endocrine glands and define the vascular changes in aging and endocrine disorders.
Collapse
Affiliation(s)
| | | | - Anjali P. Kusumbe
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
| |
Collapse
|
31
|
Bhusal A, Lee WH, Suk K. Lipocalin-2 in Diabetic Complications of the Nervous System: Physiology, Pathology, and Beyond. Front Physiol 2021; 12:638112. [PMID: 33613327 PMCID: PMC7892766 DOI: 10.3389/fphys.2021.638112] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 01/19/2021] [Indexed: 01/04/2023] Open
Abstract
Lipocalin-2 (LCN2) is a 25 kDa secreted protein that belongs to the family of lipocalins, a group of transporters of small hydrophobic molecules such as iron, fatty acids, steroids, and lipopolysaccharide in circulation. LCN2 was previously found to be involved in iron delivery, pointing toward a potential role for LCN2 in immunity. This idea was further validated when LCN2 was found to limit bacterial growth during infections in mice by sequestering iron-laden siderophores. Recently, LCN2 was also identified as a critical regulator of energy metabolism, glucose and lipid homeostasis, and insulin function. Furthermore, studies using Lcn2 knockout mice suggest an important role for LCN2 in several biobehavioral responses, including cognition, emotion, anxiety, and feeding behavior. Owing to its expression and influence on multiple metabolic and neurological functions, there has emerged a great deal of interest in the study of relationships between LCN2 and neurometabolic complications. Thorough investigation has demonstrated that LCN2 is involved in several neurodegenerative diseases, while more recent studies have shown that LCN2 is also instrumental for the progression of diabetic complications like encephalopathy and peripheral neuropathy. Preliminary findings have shown that LCN2 is also a promising drug target and diagnostic marker for the treatment of neuropathic complications from diabetes. In particular, future translational research related to LCN2, such as the development of small-molecule inhibitors or neutralizing antibodies against LCN2, appears essential for exploring its potential as a therapeutic target.
Collapse
Affiliation(s)
- Anup Bhusal
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, South Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, South Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, South Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, South Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, South Korea
| |
Collapse
|