1
|
Gallerand A, Caillot Z, Terekhova M, Castiglione A, Leporati L, Giacchero M, Pilot T, Chang M, Dolfi B, Zair FN, Goës E, Bennetot A, Mlamla Z, Mass E, Ginhoux F, Voehringer D, Mack M, Dombrowicz D, Williams JW, Masson D, Artyomov MN, Bertola A, Ivanov S. CD226 + adipose tissue macrophages arise from MDP-derived monocytes and regulate lipid metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626330. [PMID: 39677815 PMCID: PMC11642898 DOI: 10.1101/2024.12.03.626330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Macrophages are innate immune cells present in all tissues, in which they participate in immune responses and maintenance of tissue homeostasis. They develop either from embryonic precursors or from circulating monocytes, and their functions are in part dictated by their origin. We previously observed robust monocyte recruitment and contribution to the macrophage pool in brown adipose tissue. In particular, monocytes were predicted to give rise to two phenotypically distinct macrophage subsets identifiable by CD206 or CD226 expression. In the present study, we investigated monocyte differentiation pathways in brown adipose tissue and the function of monocyte-derived macrophages. We found that bone marrow monocytes highly contributed to the CD226 + macrophage population while the CD206 + population contained mainly yolk sac-derived cells. Fate mapping analysis revealed a low contribution of GMP- and a high contribution of MDP-derived monocytes to the CD226 + macrophage subset. Importantly, adoptive transfer experiments demonstrate that MDP- but not GMP-derived monocytes are pre-conditioned to give rise to CD226 + macrophages. Using meta-analysis of single cell RNA-sequencing data, we found that MDP-derived CD226 + macrophages were present in several tissues including peritoneal cavity, adrenal glands and all adipose depots, with a particular enrichment in beige and brown fat. A similar macrophage subset was identified in humans. Functionally, while depletion of CD206 + macrophages using anti-CD115 blocking antibodies led to decreased adipose triglyceride content, genetic depletion of CD226 + macrophages caused the opposite phenotype. We thus identify CD226 + MDP-derived macrophages as a new myeloid cell type conserved across tissues and tied to lipid metabolism homeostasis.
Collapse
|
2
|
Gallerand A, Han J, Ivanov S, Randolph GJ. Mouse and human macrophages and their roles in cardiovascular health and disease. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1424-1437. [PMID: 39604762 DOI: 10.1038/s44161-024-00580-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/11/2024] [Indexed: 11/29/2024]
Abstract
The past 15 years have witnessed a leap in understanding the life cycle, gene expression profiles, origins and functions of mouse macrophages in many tissues, including macrophages of the artery wall and heart that have critical roles in cardiovascular health. Here, we review the phenotypical and functional diversity of macrophage populations in multiple organs and discuss the roles that proliferation, survival, and recruitment and replenishment from monocytes have in maintaining macrophages in homeostasis and inflammatory states such as atherosclerosis and myocardial infarction. We also introduce emerging data that better characterize the life cycle and phenotypic profiles of human macrophages. We discuss the similarities and differences between murine and human macrophages, raising the possibility that tissue-resident macrophages in humans may rely more on bone marrow-derived monocytes than in mouse.
Collapse
Affiliation(s)
- Alexandre Gallerand
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jichang Han
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
3
|
Drüeke TB, Alhenc-Gelas F. A new mechanism in adrenal control of aldosterone secretion involving the macrophage and VEGF. Kidney Int 2024; 106:1011-1014. [PMID: 39299499 DOI: 10.1016/j.kint.2024.08.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024]
Affiliation(s)
- Tilman B Drüeke
- Inserm Unit 1018, Team 5, CESP, Hôpital Paul Brousse, Paris-Sud University (UPS) and Versailles Saint-Quentin-en-Yvelines University (Paris-Ile-de-France-Ouest University, UVSQ), Villejuif, France.
| | - François Alhenc-Gelas
- Inserm U 1038, Centre de recherche des Cordeliers, Paris-Cité University, Sorbonne-University, Paris, France
| |
Collapse
|
4
|
Gallerand A, Dolfi B, Stunault MI, Caillot Z, Castiglione A, Strazzulla A, Chen C, Heo GS, Luehmann H, Batoul F, Vaillant N, Dumont A, Pilot T, Merlin J, Zair FN, Gilleron J, Bertola A, Carmeliet P, Williams JW, Arguello RJ, Masson D, Dombrowicz D, Yvan-Charvet L, Doyen D, Haschemi A, Liu Y, Guinamard RR, Ivanov S. Glucose metabolism controls monocyte homeostasis and migration but has no impact on atherosclerosis development in mice. Nat Commun 2024; 15:9027. [PMID: 39424804 PMCID: PMC11489573 DOI: 10.1038/s41467-024-53267-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
Monocytes directly contribute to atherosclerosis development by their recruitment to plaques in which they differentiate into macrophages. In the present study, we ask how modulating monocyte glucose metabolism could affect their homeostasis and their impact on atherosclerosis. Here we investigate how circulating metabolites control monocyte behavior in blood, bone marrow and peripheral tissues of mice. We find that serum glucose concentrations correlate with monocyte numbers. In diet-restricted mice, monocytes fail to metabolically reprogram from glycolysis to fatty acid oxidation, leading to reduced monocyte numbers in the blood. Mechanistically, Glut1-dependent glucose metabolism helps maintain CD115 membrane expression on monocytes and their progenitors, and regulates monocyte migratory capacity by modulating CCR2 expression. Results from genetic models and pharmacological inhibitors further depict the relative contribution of different metabolic pathways to the regulation of CD115 and CCR2 expression. Meanwhile, Glut1 inhibition does not impact atherosclerotic plaque development in mouse models despite dramatically reducing blood monocyte numbers, potentially due to the remaining monocytes having increased migratory capacity. Together, these data emphasize the role of glucose uptake and intracellular glucose metabolism in controlling monocyte homeostasis and functions.
Collapse
Affiliation(s)
- Alexandre Gallerand
- Université Côte d'Azur, CNRS, LP2M, Nice, France.
- Université Côte d'Azur, INSERM, C3M, Nice, France.
| | - Bastien Dolfi
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | | | - Alexia Castiglione
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | - Chuqiao Chen
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Gyu Seong Heo
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Flora Batoul
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | | | - Thomas Pilot
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France
| | | | | | | | | | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute (LKI), KU Leuven, Leuven, 3000, Belgium
| | - Jesse W Williams
- Center for Immunology, Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Rafael J Arguello
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - David Masson
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France
| | - David Dombrowicz
- Univ.Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | | | - Denis Doyen
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Médecine Intensive Réanimation, Hôpital Pasteur, CHU de Nice, Nice, France
| | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rodolphe R Guinamard
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | - Stoyan Ivanov
- Université Côte d'Azur, CNRS, LP2M, Nice, France.
- Université Côte d'Azur, INSERM, C3M, Nice, France.
| |
Collapse
|
5
|
Paparini DE, Grasso E, Aguilera F, Arslanian MA, Lella V, Lara B, Schafir A, Gori S, Merech F, Hauk V, Schuster C, Martí M, Meller C, Ramhorst R, Vota D, Leirós CP. Sex-specific phenotypical, functional and metabolic profiles of human term placenta macrophages. Biol Sex Differ 2024; 15:80. [PMID: 39420346 PMCID: PMC11484421 DOI: 10.1186/s13293-024-00652-w] [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: 04/30/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Placental macrophages, Hofbauer cells (HBC) are the only fetal immune cell population within the stroma of healthy placenta along pregnancy. They are central players in maintaining immune tolerance during pregnancy. Immunometabolism emerged a few years ago as a new field that integrates cellular metabolism with immune responses, however, the immunometabolism of HBC has not been explored yet. Here we studied the sex-specific differences in the phenotypic, functional and immunometabolic profile of HBC. METHODS HBC were isolated from human term placentas (N = 31, 16 from male and 15 female neonates). Ex vivo assays were carried out to assess active metabolic and endoplasmic reticulum stress pathways by flow cytometry, confocal microscopy, gene expression and in silico approaches. RESULTS HBC from female placentas displayed a stronger M2 phenotype accompanied by high rates of efferocytosis majorly sustained on lipid metabolism. On the other hand, male HBC expressed a weaker M2 phenotype with higher glycolytic metabolism. LPS stimulation reinforced the glycolytic metabolism in male but not in female HBC. Physiological endoplasmic reticulum stress activates IRE-1 differently, since its pharmacological inhibition increased lipid mobilization, accumulation and efferocytosis only in female HBC. Moreover, differential sex-associated pathways accompanying the phenotypic and functional profiles of HBC appeared related to the placental villi environment. CONCLUSIONS These results support sex-associated effects on the immunometabolism of the HBC and adds another layer of complexity to the intricate maternal-fetal immune interaction.
Collapse
Affiliation(s)
- Daniel E Paparini
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| | - Esteban Grasso
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Franco Aguilera
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Bioinformatic Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | | - Victoria Lella
- Obstetric Service, Hospital Italiano, Buenos Aires, Argentina
| | - Brenda Lara
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ana Schafir
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Soledad Gori
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Fátima Merech
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Vanesa Hauk
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Claudio Schuster
- Bioinformatic Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Marcelo Martí
- Bioinformatic Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Cesar Meller
- Obstetric Service, Hospital Italiano, Buenos Aires, Argentina
| | - Rosanna Ramhorst
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Daiana Vota
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Claudia Pérez Leirós
- Immunopharmacology Laboratory, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
6
|
Xu Y, Hillman H, Chang M, Ivanov S, Williams JW. Identification of conserved and tissue-restricted transcriptional profiles for lipid associated macrophages (LAMs). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614807. [PMID: 39386558 PMCID: PMC11463620 DOI: 10.1101/2024.09.24.614807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Macrophages are essential immune cells present in all tissues, and are vital for maintaining tissue homeostasis, immune surveillance, and immune responses. Considerable efforts have identified shared and tissue-specific gene programs for macrophages across organs during homeostasis. This information has dramatically enhanced our understanding of tissue-restricted macrophage programming and function. However, few studies have addressed the overlapping and tissue-specific responses of macrophage subsets following inflammatory responses. One subset of macrophages that has been observed across several studies, lipid-associated macrophages (LAMs), have gained interest due to their unique role in lipid metabolism and potential as a therapeutic target. LAMs have been associated with regulating disease outcomes in metabolically related disorders including atherosclerosis, obesity, and nonalcoholic fatty liver disease (NAFLD). In this study, we utilized single-cell RNA sequencing (scRNAseq) data to profile LAMs across multiple tissues and sterile inflammatory conditions in mice and humans. Integration of data from various disease models revealed that LAMs share a set of conserved transcriptional profiles, including Trem2 and Lpl, but also identified key sets of tissue-specific LAM gene programs. Importantly, the shared LAM markers were highly conserved with human LAM populations that also emerge in chronic inflammatory settings. Overall, this analysis provides a detailed transcriptional landscape of tissue-restricted and shared LAM gene programs and offers insights into their roles in metabolic and chronic inflammatory diseases. These data may help instruct appropriate targets for broad or tissue-restricted therapeutic interventions to modulate LAM populations in disease.
Collapse
Affiliation(s)
- Yingzheng Xu
- Center for Immunology, University of Minnesota, Minneapolis, MN USA
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN USA
| | - Hannah Hillman
- Center for Immunology, University of Minnesota, Minneapolis, MN USA
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN USA
| | - Michael Chang
- Center for Immunology, University of Minnesota, Minneapolis, MN USA
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN USA
| | | | - Jesse W. Williams
- Center for Immunology, University of Minnesota, Minneapolis, MN USA
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN USA
| |
Collapse
|
7
|
Wensveen FM, Šestan M, Polić B. The immunology of sickness metabolism. Cell Mol Immunol 2024; 21:1051-1065. [PMID: 39107476 PMCID: PMC11364700 DOI: 10.1038/s41423-024-01192-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/29/2024] [Indexed: 09/01/2024] Open
Abstract
Everyone knows that an infection can make you feel sick. Although we perceive infection-induced changes in metabolism as a pathology, they are a part of a carefully regulated process that depends on tissue-specific interactions between the immune system and organs involved in the regulation of systemic homeostasis. Immune-mediated changes in homeostatic parameters lead to altered production and uptake of nutrients in circulation, which modifies the metabolic rate of key organs. This is what we experience as being sick. The purpose of sickness metabolism is to generate a metabolic environment in which the body is optimally able to fight infection while denying vital nutrients for the replication of pathogens. Sickness metabolism depends on tissue-specific immune cells, which mediate responses tailored to the nature and magnitude of the threat. As an infection increases in severity, so do the number and type of immune cells involved and the level to which organs are affected, which dictates the degree to which we feel sick. Interestingly, many alterations associated with metabolic disease appear to overlap with immune-mediated changes observed following infection. Targeting processes involving tissue-specific interactions between activated immune cells and metabolic organs therefore holds great potential for treating both people with severe infection and those with metabolic disease. In this review, we will discuss how the immune system communicates in situ with organs involved in the regulation of homeostasis and how this communication is impacted by infection.
Collapse
Affiliation(s)
| | - Marko Šestan
- University of Rijeka Faculty of Medicine, Rijeka, Croatia
| | - Bojan Polić
- University of Rijeka Faculty of Medicine, Rijeka, Croatia
| |
Collapse
|
8
|
Leek C, Cantu A, Sonti S, Gutierrez MC, Eldredge L, Sajti E, Xu HN, Lingappan K. Role of sex as a biological variable in neonatal alveolar macrophages. Redox Biol 2024; 75:103296. [PMID: 39098263 PMCID: PMC11345582 DOI: 10.1016/j.redox.2024.103296] [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: 05/29/2024] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024] Open
Abstract
The lung macrophages play a crucial role in health and disease. Sexual dimorphism significantly impacts the phenotype and function of tissue-resident macrophages. The primary mechanisms responsible for sexually dimorphic outcomes in bronchopulmonary dysplasia (BPD) remain unidentified. We tested the hypothesis that biological sex plays a crucial role in the transcriptional state of alveolar macrophages, using neonatal murine hyperoxia-induced lung injury as a relevant model for human BPD. The effects of neonatal hyperoxia exposure (95 % FiO2, PND1-5: saccular stage) on the lung myeloid cells acutely after injury and during normoxic recovery were measured. Alveolar macrophages (AM) from room air- and hyperoxia exposed from male and female neonatal murine lungs were subjected to bulk-RNA Sequencing. AMs are significantly depleted in the hyperoxia-exposed lung acutely after injury, with subsequent recovery in both sexes. The transcriptome of the alveolar macrophages is impacted by neonatal hyperoxia exposure and by sex as a biological variable. Pathways related to DNA damage and interferon-signaling were positively enriched in female AMs. Metabolic pathways related to glucose and carbohydrate metabolism were positively enriched in the male AMs, while oxidative phosphorylation was negatively enriched. These pathways were shared with monocytes and airway macrophages from intubated male and female human premature neonates.
Collapse
Affiliation(s)
- Connor Leek
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Abiud Cantu
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Shilpa Sonti
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Manuel Cantu Gutierrez
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Laurie Eldredge
- Department of Pediatrics, Division of Pediatric Pulmonology, University of Washington School of Medicine, Seattle Children's Hospital, WA, USA
| | - Eniko Sajti
- Department of Pediatrics, Division of Neonatology, University of California San Diego, San Diego, CA, USA
| | - He N Xu
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Krithika Lingappan
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA.
| |
Collapse
|
9
|
Altieri B, Secener AK, Sai S, Fischer C, Sbiera S, Arampatzi P, Kircher S, Herterich S, Landwehr L, Vitcetz SN, Braeuning C, Fassnacht M, Ronchi CL, Sauer S. Single-nucleus and spatial transcriptome reveal adrenal homeostasis in normal and tumoural adrenal glands. Clin Transl Med 2024; 14:e1798. [PMID: 39167619 PMCID: PMC11338279 DOI: 10.1002/ctm2.1798] [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: 11/21/2023] [Revised: 07/11/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
The human adrenal gland is a complex endocrine tissue. Studies on adrenal renewal have been limited to animal models or human foetuses. Enhancing our understanding of adult human adrenal homeostasis is crucial for gaining insights into the pathogenesis of adrenal diseases, such as adrenocortical tumours. Here, we present a comprehensive cellular genomics analysis of the adult human normal adrenal gland, combining single-nuclei RNA sequencing and spatial transcriptome data to reconstruct adrenal gland homeostasis. As expected, we identified primary cells of the various zones of the adrenal cortex and medulla, but we also uncovered additional cell types. They constitute the adrenal microenvironment, including immune cells, mostly composed of a large population of M2 macrophages, and new cell populations, including different subpopulations of vascular-endothelial cells and cortical-neuroendocrine cells. Utilizing spatial transcriptome and pseudotime trajectory analysis, we support evidence of the centripetal dynamics of adrenocortical cell maintenance and the essential role played by Wnt/β-catenin, sonic hedgehog, and fibroblast growth factor pathways in the adult adrenocortical homeostasis. Furthermore, we compared single-nuclei transcriptional profiles obtained from six healthy adrenal glands and twelve adrenocortical adenomas. This analysis unveiled a notable heterogeneity in cell populations within the adenoma samples. In addition, we identified six distinct adenoma-specific clusters, each with varying distributions based on steroid profiles and tumour mutational status. Overall, our results provide novel insights into adrenal homeostasis and molecular mechanisms potentially underlying early adrenocortical tumorigenesis and/or autonomous steroid secretion. Our cell atlas represents a powerful resource to investigate other adrenal-related pathologies.
Collapse
Affiliation(s)
- Barbara Altieri
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
| | - A. Kerim Secener
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
- Department of BiologyChemistry and PharmacyInstitute of BiochemistryFree University BerlinBerlinGermany
| | - Somesh Sai
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
- Department of BiologyChemistry and PharmacyInstitute of BiochemistryFree University BerlinBerlinGermany
| | - Cornelius Fischer
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Silviu Sbiera
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
| | | | - Stefan Kircher
- Institute of PathologyUniversity of WürzburgWürzburgGermany
| | | | - Laura‐Sophie Landwehr
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
| | - Sarah N. Vitcetz
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | | | - Martin Fassnacht
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
- Central Laboratory University Hospital WürzburgWürzburgGermany
| | - Cristina L. Ronchi
- Division of Endocrinology and DiabetesDepartment of Internal Medicine IUniversity HospitalUniversity of WürzburgWürzburgGermany
- Institute of Metabolism and System ResearchUniversity of BirminghamEdgabston, BirminghamUK
| | - Sascha Sauer
- Max Delbrück Center for Molecular MedicineBerlinGermany
- Berlin Institute of HealthBerlinGermany
- Core Unit SysMedUniversity of WürzburgWürzburgGermany
| |
Collapse
|
10
|
Dolfi B, Gallerand A, Caillot Z, Castiglione A, Zair FN, Leporati L, Giacchero M, Goës E, Strazzulla A, Dombrowicz D, Guinamard RR, Bertola A, Ivanov S. Sex-specific impact of psychosocial stress on hematopoiesis and blood leukocytes. Eur J Immunol 2024; 54:e2350851. [PMID: 38803021 DOI: 10.1002/eji.202350851] [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: 10/20/2023] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024]
Abstract
Stress exposure has been shown to modulate innate and adaptive immune responses. Indeed, stress favors myelopoiesis and monocyte generation and contributes to cardiovascular disease development. As sex hormones regulate innate and adaptive immune responses, we decided to investigate whether stress exposure leads to a different immune response in female and male mice. Our data demonstrated that psychosocial stressinduced neutrophilia in male, but not female mice. Importantly, we identified that B-cell numbers were reduced in female, but not male mice upon exposure to stress. Thus, our study revealed that the stress-induced immune alterations are sex-dependent, and this is an important feature to consider for future investigations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Eloïse Goës
- Université Côte d'Azur, CNRS, LP2M, Nice, France
| | | | - David Dombrowicz
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | | | | | | |
Collapse
|
11
|
Fan Z, Karakone M, Nagarajan S, Nagy N, Mildenberger W, Petrova E, Hinte LC, Bijnen M, Häne P, Nelius E, Chen J, Ferapontova I, von Meyenn F, Trepiccione F, Berber M, Ribas DP, Eichmann A, Zennaro MC, Takeda N, Fischer JW, Spyroglou A, Reincke M, Beuschlein F, Loffing J, Greter M, Stockmann C. Macrophages preserve endothelial cell specialization in the adrenal gland to modulate aldosterone secretion and blood pressure. Cell Rep 2024; 43:114395. [PMID: 38941187 DOI: 10.1016/j.celrep.2024.114395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/22/2024] [Accepted: 06/07/2024] [Indexed: 06/30/2024] Open
Abstract
Macrophages play crucial roles in organ-specific functions and homeostasis. In the adrenal gland, macrophages closely associate with sinusoidal capillaries in the aldosterone-producing zona glomerulosa. We demonstrate that macrophages preserve capillary specialization and modulate aldosterone secretion. Using macrophage-specific deletion of VEGF-A, single-cell transcriptomics, and functional phenotyping, we found that the loss of VEGF-A depletes PLVAP+ fenestrated endothelial cells in the zona glomerulosa, leading to increased basement membrane collagen IV deposition and subendothelial fibrosis. This results in increased aldosterone secretion, called "haptosecretagogue" signaling. Human aldosterone-producing adenomas also show capillary rarefaction and basement membrane thickening. Mice with myeloid cell-specific VEGF-A deletion exhibit elevated serum aldosterone, hypokalemia, and hypertension, mimicking primary aldosteronism. These findings underscore macrophage-to-endothelial cell signaling as essential for endothelial cell specialization, adrenal gland function, and blood pressure regulation, with broader implications for other endocrine organs.
Collapse
Affiliation(s)
- Zheng Fan
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland.
| | - Mara Karakone
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | | | - Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wiebke Mildenberger
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Ekaterina Petrova
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Laura Catharina Hinte
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Mitchell Bijnen
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Philipp Häne
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Eric Nelius
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - Jing Chen
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - Irina Ferapontova
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - Ferdinand von Meyenn
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Francesco Trepiccione
- Department of Translational Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Mesut Berber
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - David Penton Ribas
- Electrophysiology Facility (e-phac), Department of Molecular Life Sciences, University of Zurich (UZH), 8057 Zürich, Switzerland
| | - Anne Eichmann
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Norihiko Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Jens W Fischer
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Ariadni Spyroglou
- Klinik für Endokrinologie, Diabetologie, und Klinische Ernährung, UniversitätsSpital Zürich (USZ) and UZH, Raemistrasse 100, 8091 Zurich, Switzerland; Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Martin Reincke
- Klinik für Endokrinologie, Diabetologie, und Klinische Ernährung, UniversitätsSpital Zürich (USZ) and UZH, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Felix Beuschlein
- Klinik für Endokrinologie, Diabetologie, und Klinische Ernährung, UniversitätsSpital Zürich (USZ) and UZH, Raemistrasse 100, 8091 Zurich, Switzerland; Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Johannes Loffing
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland
| | - Melanie Greter
- University of Zurich, Institute for Experimental Immunology, 8057 Zurich, Switzerland
| | - Christian Stockmann
- University of Zurich, Institute of Anatomy, 8057 Zurich, Switzerland; INSERM U970, Paris Cardiovascular Research Center, Paris, France.
| |
Collapse
|
12
|
Xu Y, Patterson MT, Dolfi B, Zhu A, Bertola A, Schrank PR, Gallerand A, Kennedy AE, Hillman H, Dinh L, Shekhar S, Tollison S, Bold TD, Ivanov S, Williams JW. Adrenal gland macrophages regulate glucocorticoid production through Trem2 and TGF-β. JCI Insight 2024; 9:e174746. [PMID: 38869957 PMCID: PMC11383592 DOI: 10.1172/jci.insight.174746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 06/07/2024] [Indexed: 06/15/2024] Open
Abstract
Glucocorticoid synthesis by adrenal glands (AGs) is regulated by the hypothalamic-pituitary-adrenal axis to facilitate stress responses when the host is exposed to stimuli. Recent studies implicate macrophages as potential steroidogenic regulators, but the molecular mechanisms by which AG macrophages exert such influence remain unclear. In this study, we investigated the role of AG macrophages in response to cold challenge or atherosclerotic inflammation as physiologic models of acute or chronic stress. Using single-cell RNA sequencing, we observed dynamic AG macrophage polarization toward classical activation and lipid-associated phenotypes following acute or chronic stimulation. Among transcriptional alterations induced in macrophages, triggering receptor expressed on myeloid cells 2 (Trem2) was highlighted because of its upregulation following stress. Conditional deletion of macrophage Trem2 revealed a protective role in stress responses. Mechanistically, Trem2 deletion led to increased AG macrophage death, abolished the TGF-β-producing capacity of AG macrophages, and resulted in enhanced glucocorticoid production. In addition, enhanced glucocorticoid production was replicated by blockade of TGF-β signaling. Together, these observations suggest that AG macrophages restrict steroidogenesis through Trem2 and TGF-β, which opens potential avenues for immunotherapeutic interventions to resolve stress-related disorders.
Collapse
Affiliation(s)
- Yingzheng Xu
- Center for Immunology and
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael T Patterson
- Center for Immunology and
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Alisha Zhu
- Center for Immunology and
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Patricia R Schrank
- Center for Immunology and
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Ainsley E Kennedy
- Center for Immunology and
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Hannah Hillman
- Center for Immunology and
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lynn Dinh
- Center for Immunology and
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sia Shekhar
- Center for Immunology and
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Samuel Tollison
- Center for Immunology and
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Tyler D Bold
- Center for Immunology and
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Jesse W Williams
- Center for Immunology and
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
13
|
Iwahashi N, Umakoshi H, Fujita M, Fukumoto T, Ogasawara T, Yokomoto-Umakoshi M, Kaneko H, Nakao H, Kawamura N, Uchida N, Matsuda Y, Sakamoto R, Seki M, Suzuki Y, Nakatani K, Izumi Y, Bamba T, Oda Y, Ogawa Y. Single-cell and spatial transcriptomics analysis of human adrenal aging. Mol Metab 2024; 84:101954. [PMID: 38718896 PMCID: PMC11101872 DOI: 10.1016/j.molmet.2024.101954] [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: 01/09/2024] [Revised: 03/30/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
OBJECTIVE The human adrenal cortex comprises three functionally and structurally distinct layers that produce layer-specific steroid hormones. With aging, the human adrenal cortex undergoes functional and structural alteration or "adrenal aging", leading to the unbalanced production of steroid hormones. Given the marked species differences in adrenal biology, the underlying mechanisms of human adrenal aging have not been sufficiently studied. This study was designed to elucidate the mechanisms linking the functional and structural alterations of the human adrenal cortex. METHODS We conducted single-cell RNA sequencing and spatial transcriptomics analysis of the aged human adrenal cortex. RESULTS The data of this study suggest that the layer-specific alterations of multiple signaling pathways underlie the abnormal layered structure and layer-specific changes in steroidogenic cells. We also highlighted that macrophages mediate age-related adrenocortical cell inflammation and senescence. CONCLUSIONS This study is the first detailed analysis of the aged human adrenal cortex at single-cell resolution and helps to elucidate the mechanism of human adrenal aging, thereby leading to a better understanding of the pathophysiology of age-related disorders associated with adrenal aging.
Collapse
Affiliation(s)
- Norifusa Iwahashi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hironobu Umakoshi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Masamichi Fujita
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tazuru Fukumoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsuki Ogasawara
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Maki Yokomoto-Umakoshi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroki Kaneko
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroshi Nakao
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Namiko Kawamura
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naohiro Uchida
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yayoi Matsuda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryuichi Sakamoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Kohta Nakatani
- Division of Metabolomics/Mass Spectrometry Center, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics/Mass Spectrometry Center, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takeshi Bamba
- Division of Metabolomics/Mass Spectrometry Center, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
14
|
Niepoth N, Merritt JR, Uminski M, Lei E, Esquibies VS, Bando IB, Hernandez K, Gebhardt C, Wacker SA, Lutzu S, Poudel A, Soma KK, Rudolph S, Bendesky A. Evolution of a novel adrenal cell type that promotes parental care. Nature 2024; 629:1082-1090. [PMID: 38750354 PMCID: PMC11329292 DOI: 10.1038/s41586-024-07423-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/15/2024] [Indexed: 05/25/2024]
Abstract
Cell types with specialized functions fundamentally regulate animal behaviour, and yet the genetic mechanisms that underlie the emergence of novel cell types and their consequences for behaviour are not well understood1. Here we show that the monogamous oldfield mouse (Peromyscus polionotus) has recently evolved a novel cell type in the adrenal gland that expresses the enzyme AKR1C18, which converts progesterone into 20α-hydroxyprogesterone. We then demonstrate that 20α-hydroxyprogesterone is more abundant in oldfield mice, where it induces monogamous-typical parental behaviours, than in the closely related promiscuous deer mice (Peromyscus maniculatus). Using quantitative trait locus mapping in a cross between these species, we ultimately find interspecific genetic variation that drives expression of the nuclear protein GADD45A and the glycoprotein tenascin N, which contribute to the emergence and function of this cell type in oldfield mice. Our results provide an example by which the recent evolution of a new cell type in a gland outside the brain contributes to the evolution of social behaviour.
Collapse
Affiliation(s)
- Natalie Niepoth
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Ecology, Evolution and Environmental Biology, Columbia University, New York, NY, USA
| | - Jennifer R Merritt
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Ecology, Evolution and Environmental Biology, Columbia University, New York, NY, USA
| | - Michelle Uminski
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Ecology, Evolution and Environmental Biology, Columbia University, New York, NY, USA
| | - Emily Lei
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Ecology, Evolution and Environmental Biology, Columbia University, New York, NY, USA
| | - Victoria S Esquibies
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Ecology, Evolution and Environmental Biology, Columbia University, New York, NY, USA
| | - Ina B Bando
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Ecology, Evolution and Environmental Biology, Columbia University, New York, NY, USA
| | - Kimberly Hernandez
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Ecology, Evolution and Environmental Biology, Columbia University, New York, NY, USA
| | - Christoph Gebhardt
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Ecology, Evolution and Environmental Biology, Columbia University, New York, NY, USA
| | - Sarah A Wacker
- Department of Chemistry and Biochemistry, Manhattan College, New York, NY, USA
| | - Stefano Lutzu
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, NY, USA
| | - Asmita Poudel
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kiran K Soma
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephanie Rudolph
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, NY, USA
| | - Andres Bendesky
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA.
- Department of Ecology, Evolution and Environmental Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
15
|
Zhao J, Andreev I, Silva HM. Resident tissue macrophages: Key coordinators of tissue homeostasis beyond immunity. Sci Immunol 2024; 9:eadd1967. [PMID: 38608039 DOI: 10.1126/sciimmunol.add1967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 03/18/2024] [Indexed: 04/14/2024]
Abstract
Resident tissue macrophages (RTMs) encompass a highly diverse set of cells abundantly present in every tissue and organ. RTMs are recognized as central players in innate immune responses, and more recently their importance beyond host defense has started to be highlighted. Despite sharing a universal name and several canonical markers, RTMs perform remarkably specialized activities tailored to sustain critical homeostatic functions of the organs they reside in. These cells can mediate neuronal communication, participate in metabolic pathways, and secrete growth factors. In this Review, we summarize how the division of labor among different RTM subsets helps support tissue homeostasis. We discuss how the local microenvironment influences the development of RTMs, the molecular processes they support, and how dysregulation of RTMs can lead to disease. Last, we highlight both the similarities and tissue-specific distinctions of key RTM subsets, aiming to coalesce recent classifications and perspectives into a unified view.
Collapse
Affiliation(s)
- Jia Zhao
- Laboratory of Immunophysiology, Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ilya Andreev
- Laboratory of Immunophysiology, Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hernandez Moura Silva
- Laboratory of Immunophysiology, Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| |
Collapse
|
16
|
Gao Y, Tian X, Zhang X, Milebe Nkoua GD, Chen F, Liu Y, Chai Y. The roles of tissue-resident macrophages in sepsis-associated organ dysfunction. Heliyon 2023; 9:e21391. [PMID: 38027963 PMCID: PMC10643296 DOI: 10.1016/j.heliyon.2023.e21391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/13/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis, a syndrome caused by a dysregulated host response to infection and characterized by life-threatening organ dysfunction, particularly septic shock and sepsis-associated organ dysfunction (SAOD), is a medical emergency associated with high morbidity, high mortality, and long-term sequelae. Tissue-resident macrophages (TRMs) are a subpopulation of macrophages derived primarily from yolk sac progenitors and fetal liver during embryogenesis, located primarily in non-lymphoid tissues in adulthood, capable of local self-renewal independent of hematopoiesis, and developmentally and functionally restricted to the non-lymphoid organs in which they reside. TRMs are the first line of defense against life-threatening conditions such as sepsis, tumor growth, traumatic-associated organ injury, and surgical-associated injury. In the context of sepsis, TRMs can be considered as angels or demons involved in organ injury. Our proposal is that sepsis, septic shock, and SAOD can be attenuated by modulating TRMs in different organs. This review summarizes the pathophysiological mechanisms of TRMs in different organs or tissues involved in the development and progression of sepsis.
Collapse
Affiliation(s)
- Yulei Gao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
- Department of Emergency Medicine, China-Congo Friendship Hospital, Brazzaville, 999059, P. R. Congo
| | - Xin Tian
- Department of Medical Research, Beijing Qiansong Technology Development Company, Beijing, 100193, P. R. China
- Department of Medical Research, Sen Sho Ka Gi Company, Inba-gun, Chiba, 285-0905, Japan
| | - Xiang Zhang
- Department of Emergency Medicine, Rizhao People's Hospital of Shandong Province, Rizhao, 276825, P. R. China
| | | | - Fang Chen
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
| | - Yancun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
| | - Yanfen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
| |
Collapse
|
17
|
Warde KM, Smith LJ, Basham KJ. Age-related Changes in the Adrenal Cortex: Insights and Implications. J Endocr Soc 2023; 7:bvad097. [PMID: 37564884 PMCID: PMC10410302 DOI: 10.1210/jendso/bvad097] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Indexed: 08/12/2023] Open
Abstract
Aging is characterized by a gradual decline in physiological function. This process affects all organs including the adrenal cortex, which normally functions to produce essential steroid hormones including mineralocorticoids, glucocorticoids, and androgens. With increasing age, features such as reduced adrenal cortex size, altered zonation, and increased myeloid immune cell infiltration substantially alter the structure and function of the adrenal cortex. Many of these hallmark features of adrenal cortex aging occur both in males and females, yet are more enhanced in males. Hormonally, a substantial reduction in adrenal androgens is a key feature of aging, which is accompanied by modest changes in aldosterone and cortisol. These hormonal changes are associated with various pathological consequences including impaired immune responses, decreased bone health, and accelerated age-related diseases. One of the most notable changes with adrenal aging is the increased incidence of adrenal tumors, which is sex dimorphic with a higher prevalence in females. Increased adrenal tumorigenesis with age is likely driven by both an increase in genetic mutations as well as remodeling of the tissue microenvironment. Novel antiaging strategies offer a promising avenue to mitigate adrenal aging and alleviate age-associated pathologies, including adrenal tumors.
Collapse
Affiliation(s)
- Kate M Warde
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Lorenzo J Smith
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Kaitlin J Basham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
18
|
Patterson MT, Burrack AL, Xu Y, Hickok GH, Schmiechen ZC, Becker S, Cruz-Hinojoza E, Schrank PR, Kennedy AE, Firulyova MM, Miller EA, Zaitsev K, Williams JW, Stromnes IM. Tumor-specific CD4 T cells instruct monocyte fate in pancreatic ductal adenocarcinoma. Cell Rep 2023; 42:112732. [PMID: 37402168 PMCID: PMC10448358 DOI: 10.1016/j.celrep.2023.112732] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 04/21/2023] [Accepted: 06/16/2023] [Indexed: 07/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) orchestrates a suppressive tumor microenvironment that fosters immunotherapy resistance. Tumor-associated macrophages (TAMs) are the principal immune cell infiltrating PDA and are heterogeneous. Here, by employing macrophage fate-mapping approaches and single-cell RNA sequencing, we show that monocytes give rise to most macrophage subsets in PDA. Tumor-specific CD4, but not CD8, T cells promote monocyte differentiation into MHCIIhi anti-tumor macrophages. By conditional major histocompatibility complex (MHC) class II deletion on monocyte-derived macrophages, we show that tumor antigen presentation is required for instructing monocyte differentiation into anti-tumor macrophages, promoting Th1 cells, abrogating Treg cells, and mitigating CD8 T cell exhaustion. Non-redundant IFNγ and CD40 promote MHCIIhi anti-tumor macrophages. Intratumoral monocytes adopt a pro-tumor fate indistinguishable from that of tissue-resident macrophages following loss of macrophage MHC class II or tumor-specific CD4 T cells. Thus, tumor antigen presentation by macrophages to CD4 T cells dictates TAM fate and is a major determinant of macrophage heterogeneity in cancer.
Collapse
Affiliation(s)
- Michael T Patterson
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Adam L Burrack
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Yingzheng Xu
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Grant H Hickok
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Zoe C Schmiechen
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Samuel Becker
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Eduardo Cruz-Hinojoza
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Patricia R Schrank
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Ainsley E Kennedy
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Maria M Firulyova
- Computer Technologies Laboratory, ITMO University, Saint-Petersburg, Russia; National Medical Research Center, Saint-Petersburg, Russia
| | - Ebony A Miller
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Konstantin Zaitsev
- Computer Technologies Laboratory, ITMO University, Saint-Petersburg, Russia
| | - Jesse W Williams
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55414, USA.
| | - Ingunn M Stromnes
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Masonic Cancer Center and University of Minnesota Medical School, Minneapolis, MN 55414, USA; Center for Genome Engineering, University of Minnesota Medical School, Minneapolis, MN 55414, USA.
| |
Collapse
|
19
|
Kang Y, Laprocina K, Zheng HS, Huang CCJ. Current insight into the transient X-zone in the adrenal gland cortex. VITAMINS AND HORMONES 2023; 124:297-339. [PMID: 38408801 PMCID: PMC11023618 DOI: 10.1016/bs.vh.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Mouse models have been widely used in the study of adrenal gland development and diseases. The X-zone is a unique structure of the mouse adrenal gland and lineage-tracing studies show that the X-zone is a remnant of the fetal adrenal cortex. Although the X-zone is considered analogous to the fetal zone in the human adrenal cortex, the functional significance of the X-zone has remained comparatively more obscure. The X-zone forms during the early postnatal stages of adrenal development and regresses later in a remarkable sexually dimorphic fashion. The formation and regression of the X-zone can be different in mice with different genetic backgrounds. Mouse models with gene mutations, hormone/chemical treatments, and/or gonadectomy can also display an aberrant development of the X-zone or alternatively a dysregulated X-zone regression. These models have shed light on the molecular mechanisms regulating the development and regression of these unique adrenocortical cells. This review paper briefly describes the development of the adrenal gland including the formation and regression processes of the X-zone. It also summarizes and lists mouse models that demonstrate different X-zone phenotypes.
Collapse
Affiliation(s)
- Yuan Kang
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Karly Laprocina
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Huifei Sophia Zheng
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Chen-Che Jeff Huang
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States.
| |
Collapse
|
20
|
Warde KM, Smith LJ, Liu L, Stubben CJ, Lohman BK, Willett PW, Ammer JL, Castaneda-Hernandez G, Imodoye SO, Zhang C, Jones KD, Converso-Baran K, Ekiz HA, Barry M, Clay MR, Kiseljak-Vassiliades K, Giordano TJ, Hammer GD, Basham KJ. Senescence-induced immune remodeling facilitates metastatic adrenal cancer in a sex-dimorphic manner. NATURE AGING 2023; 3:846-865. [PMID: 37231196 PMCID: PMC11534150 DOI: 10.1038/s43587-023-00420-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 04/12/2023] [Indexed: 05/27/2023]
Abstract
Aging markedly increases cancer risk, yet our mechanistic understanding of how aging influences cancer initiation is limited. Here we demonstrate that the loss of ZNRF3, an inhibitor of Wnt signaling that is frequently mutated in adrenocortical carcinoma, leads to the induction of cellular senescence that remodels the tissue microenvironment and ultimately permits metastatic adrenal cancer in old animals. The effects are sexually dimorphic, with males exhibiting earlier senescence activation and a greater innate immune response, driven in part by androgens, resulting in high myeloid cell accumulation and lower incidence of malignancy. Conversely, females present a dampened immune response and increased susceptibility to metastatic cancer. Senescence-recruited myeloid cells become depleted as tumors progress, which is recapitulated in patients in whom a low myeloid signature is associated with worse outcomes. Our study uncovers a role for myeloid cells in restraining adrenal cancer with substantial prognostic value and provides a model for interrogating pleiotropic effects of cellular senescence in cancer.
Collapse
Affiliation(s)
- Kate M Warde
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Lorenzo J Smith
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Lihua Liu
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Chris J Stubben
- Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Brian K Lohman
- Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Parker W Willett
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Julia L Ammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | | | - Sikiru O Imodoye
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Chenge Zhang
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Kara D Jones
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Kimber Converso-Baran
- Frankel Cardiovascular Center Physiology and Phenotyping Core, University of Michigan, Ann Arbor, MI, USA
| | - H Atakan Ekiz
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla Izmir, Turkey
| | - Marc Barry
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Michael R Clay
- Department of Pathology, University of Colorado School of Medicine at Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine at Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas J Giordano
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Kaitlin J Basham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
21
|
Lyraki R, Grabek A, Tison A, Weerasinghe Arachchige LC, Peitzsch M, Bechmann N, Youssef SA, de Bruin A, Bakker ERM, Claessens F, Chaboissier MC, Schedl A. Crosstalk between androgen receptor and WNT/β-catenin signaling causes sex-specific adrenocortical hyperplasia in mice. Dis Model Mech 2023; 16:dmm050053. [PMID: 37102205 PMCID: PMC10184674 DOI: 10.1242/dmm.050053] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/29/2023] [Indexed: 04/28/2023] Open
Abstract
Female bias is highly prevalent in conditions such as adrenal cortex hyperplasia and neoplasia, but the reasons behind this phenomenon are poorly understood. In this study, we show that overexpression of the secreted WNT agonist R-spondin 1 (RSPO1) leads to ectopic activation of WNT/β-catenin signaling and causes sex-specific adrenocortical hyperplasia in mice. Although female adrenals show ectopic proliferation, male adrenals display excessive immune system activation and cortical thinning. Using a combination of genetic manipulations and hormonal treatment, we show that gonadal androgens suppress ectopic proliferation in the adrenal cortex and determine the selective regulation of the WNT-related genes Axin2 and Wnt4. Notably, genetic removal of androgen receptor (AR) from adrenocortical cells restores the mitogenic effect of WNT/β-catenin signaling. This is the first demonstration that AR activity in the adrenal cortex determines susceptibility to canonical WNT signaling-induced hyperplasia.
Collapse
Affiliation(s)
- Rodanthi Lyraki
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, 06108 Nice, France
| | - Anaëlle Grabek
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, 06108 Nice, France
| | - Amélie Tison
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, 06108 Nice, France
| | | | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Sameh A. Youssef
- Dutch Molecular Pathology Center, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL, Utrecht, the Netherlands
- Janssen Research and Development, 2340 Beerse, Belgium
| | - Alain de Bruin
- Dutch Molecular Pathology Center, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL, Utrecht, the Netherlands
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 AV, Groningen, the Netherlands
| | - Elvira R. M. Bakker
- Department of Pathology, University Medical Center Utrecht, 3508 AB, Utrecht, the Netherlands
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | | | - Andreas Schedl
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, 06108 Nice, France
| |
Collapse
|
22
|
Tyczewska M, Sujka-Kordowska P, Szyszka M, Jopek K, Blatkiewicz M, Malendowicz LK, Rucinski M. Transcriptome Profile of the Rat Adrenal Gland: Parenchymal and Interstitial Cells. Int J Mol Sci 2023; 24:ijms24119159. [PMID: 37298112 DOI: 10.3390/ijms24119159] [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: 05/04/2023] [Revised: 05/16/2023] [Accepted: 05/21/2023] [Indexed: 06/12/2023] Open
Abstract
The homeostasis of the adrenal gland plays a decisive role in its proper functioning, both in non-stressful conditions and under the influence of various types of stress. This consists of interactions between all types of cells that make up the organ, including parenchymal and interstitial cells. The amount of available information on this subject in the rat adrenal glands under non-stressful conditions is insufficient; the aim of the research was to determine the expression of marker genes for rat adrenal cells depending on their location. The material for the study consisted of adrenal glands taken from intact adult male rats that were separated into appropriate zones. Transcriptome analysis by means of Affymetrix® Rat Gene 2.1 ST Array was used in the study, followed by real-time PCR validation. Expression analysis of interstitial cell marker genes revealed both the amount of expression of these genes and the zone in which they were expressed. The expression of marker genes for fibroblasts was particularly high in the cells of the ZG zone, while the highest expression of specific macrophage genes was observed in the adrenal medulla. The results of this study, especially with regard to interstitial cells, provide a so far undescribed model of marker gene expression of various cells, both in the cortex and medulla of the sexually mature rat adrenal gland. The interdependence between parenchymal and interstitial cells creates a specific microenvironment that is highly heterogeneous within the gland with respect to some of the interstitial cells. This phenomenon most likely depends on the interaction with the differentiated parenchymal cells of the cortex, as well as the medulla of the gland.
Collapse
Affiliation(s)
- Marianna Tyczewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecickiego 6 Street, 60-781 Poznan, Poland
| | - Patrycja Sujka-Kordowska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecickiego 6 Street, 60-781 Poznan, Poland
| | - Marta Szyszka
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecickiego 6 Street, 60-781 Poznan, Poland
| | - Karol Jopek
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecickiego 6 Street, 60-781 Poznan, Poland
| | - Małgorzata Blatkiewicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecickiego 6 Street, 60-781 Poznan, Poland
| | - Ludwik K Malendowicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecickiego 6 Street, 60-781 Poznan, Poland
| | - Marcin Rucinski
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecickiego 6 Street, 60-781 Poznan, Poland
| |
Collapse
|
23
|
Danzi F, Pacchiana R, Mafficini A, Scupoli MT, Scarpa A, Donadelli M, Fiore A. To metabolomics and beyond: a technological portfolio to investigate cancer metabolism. Signal Transduct Target Ther 2023; 8:137. [PMID: 36949046 PMCID: PMC10033890 DOI: 10.1038/s41392-023-01380-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 03/24/2023] Open
Abstract
Tumour cells have exquisite flexibility in reprogramming their metabolism in order to support tumour initiation, progression, metastasis and resistance to therapies. These reprogrammed activities include a complete rewiring of the bioenergetic, biosynthetic and redox status to sustain the increased energetic demand of the cells. Over the last decades, the cancer metabolism field has seen an explosion of new biochemical technologies giving more tools than ever before to navigate this complexity. Within a cell or a tissue, the metabolites constitute the direct signature of the molecular phenotype and thus their profiling has concrete clinical applications in oncology. Metabolomics and fluxomics, are key technological approaches that mainly revolutionized the field enabling researchers to have both a qualitative and mechanistic model of the biochemical activities in cancer. Furthermore, the upgrade from bulk to single-cell analysis technologies provided unprecedented opportunity to investigate cancer biology at cellular resolution allowing an in depth quantitative analysis of complex and heterogenous diseases. More recently, the advent of functional genomic screening allowed the identification of molecular pathways, cellular processes, biomarkers and novel therapeutic targets that in concert with other technologies allow patient stratification and identification of new treatment regimens. This review is intended to be a guide for researchers to cancer metabolism, highlighting current and emerging technologies, emphasizing advantages, disadvantages and applications with the potential of leading the development of innovative anti-cancer therapies.
Collapse
Affiliation(s)
- Federica Danzi
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Raffaella Pacchiana
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Andrea Mafficini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Maria T Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, Biology and Genetics Section, University of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- ARC-NET Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy.
| | - Alessandra Fiore
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| |
Collapse
|
24
|
Transgenic Mouse Models to Study the Development and Maintenance of the Adrenal Cortex. Int J Mol Sci 2022; 23:ijms232214388. [PMID: 36430866 PMCID: PMC9693478 DOI: 10.3390/ijms232214388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
The cortex of the adrenal gland is organized into concentric zones that produce distinct steroid hormones essential for body homeostasis in mammals. Mechanisms leading to the development, zonation and maintenance of the adrenal cortex are complex and have been studied since the 1800s. However, the advent of genetic manipulation and transgenic mouse models over the past 30 years has revolutionized our understanding of these mechanisms. This review lists and details the distinct Cre recombinase mouse strains available to study the adrenal cortex, and the remarkable progress total and conditional knockout mouse models have enabled us to make in our understanding of the molecular mechanisms regulating the development and maintenance of the adrenal cortex.
Collapse
|
25
|
Bertola A, Gallerand A, Ivanov S. Immune cell involvement in brown adipose tissue functions. DISCOVERY IMMUNOLOGY 2022; 1:kyac007. [PMID: 38566905 PMCID: PMC10917225 DOI: 10.1093/discim/kyac007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/15/2022] [Accepted: 10/28/2022] [Indexed: 04/04/2024]
Abstract
Brown adipose tissue (BAT) contains many immune cells. The presence of macrophages, monocytes, dendritic cells, T cells, B cells, and mast cells was documented in BAT. However, in comparison to white adipose tissue, relatively little is known on the impact of immune cells on BAT function. By directly interacting with BAT stromal cells, or by secreting pro- and anti-inflammatory mediators, immune cells modulate BAT activation and subsequently influence on adaptative thermogenesis and heat generation. In the current manuscript, we will focus on the diversity and functions of BAT immune cells.
Collapse
|