1
|
Colapietro AA, Grillo Balboa J, Ceol Retamal MN, Regueira E, Hermida GN, Cantarelli VI, Ponzio MF, Pallarés ME, Antonelli MC, Diaz SL. Infant Maltreatment Induces Early Alterations in Adrenal Glands and Stress Response in Juvenile Rat Offspring. Neurochem Res 2025; 50:108. [PMID: 40019650 DOI: 10.1007/s11064-025-04363-5] [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: 12/30/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
Chronic stressors, such as infant maltreatment during early development, can have long-lasting effects on an individual's health and stress-coping abilities across the lifespan. In this study, we applied the scarcity-adversity model (SAM) to Wistar rats from postnatal days (PND) 8 to 12 to evaluate the impact of scarcity condition on adrenal glands morphology and stress response in juvenile offspring (PND 21-35) of both sexes. Our results demonstrate that SAM exposure leads to significant alterations in the adrenal glands of offspring. Notably, the cortex-to-medulla ratio was reduced in SAM pups starting at PND 28. Morphological analysis of the adrenal cortex revealed an increased thickness of the undifferentiated zone, suggesting enhanced cell proliferation, alongside a transient reduction in the thickness of the zona fasciculata, where glucocorticoid-synthesizing cells are located. Additionally, we observed a reduction in the cytoplasmic diameter of zona fasciculata cells, coupled with an increase in the thickness of blood capillaries in this region in SAM offspring of both sexes. An increase in BrdU + cells in the cortex at PND 28 was observed in male SAM pups, but not in females. Endocrine assessments revealed a marked increase in corticosterone levels following stress induced by movement restriction in juvenile SAM offspring of both sexes. In conclusion, our findings underscore the significant impact of early-life stress on adrenal gland morphology and stress responsiveness, with potential long-term consequences for health and stress-coping mechanisms in offspring.
Collapse
Affiliation(s)
- Ailen Alba Colapietro
- Instituto de Biología Celular y Neurociencias ''Prof. E. De Robertis''. Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Biociencias, Biotecnología y Biología Traslacional, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Raúl Alfonsín S/N, Ciudad Universitaria, Pabellón 2, 2° Floor, C1428EGA, Buenos Aires, Argentina
| | - Jazmín Grillo Balboa
- Instituto de Biología Celular y Neurociencias ''Prof. E. De Robertis''. Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marianela Noemí Ceol Retamal
- Instituto de Biología Celular y Neurociencias ''Prof. E. De Robertis''. Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Eleonora Regueira
- Departamento de Biodiversidad y Biología Experimental. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gladys Noemí Hermida
- Departamento de Biodiversidad y Biología Experimental. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Verónica Inés Cantarelli
- Facultad de Ciencias Médicas, Instituto de Investigaciones en Ciencias de La Salud, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Marina Flavia Ponzio
- Facultad de Ciencias Médicas, Instituto de Investigaciones en Ciencias de La Salud, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Eugenia Pallarés
- Instituto de Biología Celular y Neurociencias ''Prof. E. De Robertis''. Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marta Cristina Antonelli
- Instituto de Biología Celular y Neurociencias ''Prof. E. De Robertis''. Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Silvina Laura Diaz
- Instituto de Biociencias, Biotecnología y Biología Traslacional, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Raúl Alfonsín S/N, Ciudad Universitaria, Pabellón 2, 2° Floor, C1428EGA, Buenos Aires, Argentina.
- Cátedra de Técnica de Bioterio. Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
2
|
Earnhardt-San AL, Baker EC, Cilkiz KZ, Cardoso RC, Ghaffari N, Long CR, Riggs PK, Randel RD, Riley DG, Welsh TH. Evaluation of Prenatal Transportation Stress on DNA Methylation (DNAm) and Gene Expression in the Hypothalamic-Pituitary-Adrenal (HPA) Axis Tissues of Mature Brahman Cows. Genes (Basel) 2025; 16:191. [PMID: 40004522 PMCID: PMC11855312 DOI: 10.3390/genes16020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/27/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The experience of prenatal stress results in various physiological disorders due to an alteration of an offspring's methylome and transcriptome. The objective of this study was to determine whether PNS affects DNA methylation (DNAm) and gene expression in the stress axis tissues of mature Brahman cows. Methods: Samples were collected from the paraventricular nucleus (PVN), anterior pituitary (PIT), and adrenal cortex (AC) of 5-year-old Brahman cows that were prenatally exposed to either transportation stress (PNS, n = 6) or were not transported (Control, n = 8). The isolated DNA and RNA samples were, respectively, used for methylation and RNA-Seq analyses. A gene ontology and KEGG pathway enrichment analysis of each data set within each sample tissue was conducted with the DAVID Functional Annotation Tool. Results: The DNAm analysis revealed 3, 64, and 99 hypomethylated and 2, 93, and 90 hypermethylated CpG sites (FDR < 0.15) within the PVN, PIT, and AC, respectively. The RNA-Seq analysis revealed 6, 25, and 5 differentially expressed genes (FDR < 0.15) in the PVN, PIT, and AC, respectively, that were up-regulated in the PNS group relative to the Control group, as well as 24 genes in the PIT that were down-regulated. Based on the enrichment analysis, several developmental and cellular processes, such as maintenance of the actin cytoskeleton, cell motility, signal transduction, neurodevelopment, and synaptic function, were potentially modulated. Conclusions: The methylome and transcriptome were altered in the stress axis tissues of mature cows that had been exposed to prenatal transportation stress. These findings are relevant to understanding how prenatal experiences may affect postnatal neurological functions.
Collapse
Affiliation(s)
- Audrey L. Earnhardt-San
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Texas A&M AgriLife Research Center, Overton, TX 75684, USA
| | - Emilie C. Baker
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Kubra Z. Cilkiz
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Rodolfo C. Cardoso
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Noushin Ghaffari
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - Charles R. Long
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - Penny K. Riggs
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Ronald D. Randel
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
- Department of Computer Science, Prairie View A&M University, Prairie View, TX 77070, USA;
| | - David G. Riley
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| | - Thomas H. Welsh
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (A.L.E.-S.); (E.C.B.); (K.Z.C.); (R.C.C.); (C.R.L.); (P.K.R.); (R.D.R.); (D.G.R.)
| |
Collapse
|
3
|
Tourigny DS, Altieri B, Secener KA, Sbiera S, Schauer MP, Arampatzi P, Herterich S, Sauer S, Fassnacht M, Ronchi CL. Cellular landscape of adrenocortical carcinoma at single-nuclei resolution. Mol Cell Endocrinol 2024; 590:112272. [PMID: 38759836 DOI: 10.1016/j.mce.2024.112272] [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: 03/11/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Adrenocortical carcinoma (ACC) is a rare yet devastating tumour of the adrenal gland with a molecular pathology that remains incompletely understood. To gain novel insights into the cellular landscape of ACC, we generated single-nuclei RNA sequencing (snRNA-seq) data sets from twelve ACC tumour samples and analysed these alongside snRNA-seq data sets from normal adrenal glands (NAGs). We find the ACC tumour microenvironment to be relatively devoid of immune cells compared to NAG tissues, consistent with known high tumour purity values for ACC as an immunologically "cold" tumour. Our analysis identifies three separate groups of ACC samples that are characterised by different relative compositions of adrenocortical cell types. These include cell populations that are specifically enriched in the most clinically aggressive and hormonally active tumours, displaying hallmarks of reorganised cell mechanobiology and dysregulated steroidogenesis, respectively. We also identified and validated a population of mitotically active adrenocortical cells that strongly overexpress genes POLQ, DIAPH3 and EZH2 to support tumour expansion alongside an LGR4+ progenitor-like or cell-of-origin candidate for adrenocortical carcinogenesis. Trajectory inference suggests the fate adopted by malignant adrenocortical cells upon differentiation is associated with the copy number or allelic balance state of the imprinted DLK1/MEG3 genomic locus, which we verified by assessing bulk tumour DNA methylation status. In conclusion, our results therefore provide new insights into the clinical and cellular heterogeneity of ACC, revealing how genetic perturbations to healthy adrenocortical renewal and zonation provide a molecular basis for disease pathogenesis.
Collapse
Affiliation(s)
- David S Tourigny
- School of Mathematics, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Barbara Altieri
- Division of Endocrinology and Diabetes, University Hospital of Würzburg, Würzburg, 97080, Germany
| | - Kerim A Secener
- Max Delbrück Center for Molecular Medicine, Berlin, 13125, Germany; Institute of Biochemistry, Department of Biology, Chemistry and Pharmacy, Free University Berlin, Berlin, 14195, Germany
| | - Silviu Sbiera
- Division of Endocrinology and Diabetes, University Hospital of Würzburg, Würzburg, 97080, Germany
| | - Marc P Schauer
- Division of Endocrinology and Diabetes, University Hospital of Würzburg, Würzburg, 97080, Germany; Center for Cellular Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, 97080, Germany
| | | | - Sabine Herterich
- Central Laboratory, University Hospital of Würzburg, Würzburg, 97080, Germany
| | - Sascha Sauer
- Max Delbrück Center for Molecular Medicine, Berlin, 13125, Germany
| | - Martin Fassnacht
- Division of Endocrinology and Diabetes, University Hospital of Würzburg, Würzburg, 97080, Germany
| | - Cristina L Ronchi
- Institute of Metabolism and System Research, University of Birmingham, Birmingham, B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, B15 2GW, UK.
| |
Collapse
|
4
|
Kremer JL, Santiago VF, Bongiovani Rodrigues F, Auricino TB, Freitas DHDO, Palmisano G, Lotfi CFP. Extracellular Matrix Protein Signatures of the Outer and Inner Zones of the Rat Adrenal Cortex. J Proteome Res 2024; 23:3418-3432. [PMID: 39018382 DOI: 10.1021/acs.jproteome.4c00071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
This study analyzes the extracellular matrix (ECM) signatures of the outer (OF = capsule + subcapsular + zona glomerulosa cells) and inner fractions (IF = zona fasciculata cells) of the rat adrenal cortex, which comprise two distinct microenvironment niches. Proteomic profiles of decellularized OF and IF samples, male and female rats, identified 252 proteins, with 32 classified as ECM-component and ECM-related. Among these, 25 proteins were differentially regulated: 17 more abundant in OF, including Col1a1, Col1a2, Col6a1, Col6a2, Col6a3, Col12a1, Col14a1, Lama5, Lamb2, Lamc1, Eln, Emilin, Fbln5, Fbn1, Fbn2, Nid1, and Ltbp4, and eight more abundant in IF, including Col4a1, Col4a2, Lama2, Lama4, Lamb1, Fn1, Hspg2, and Ecm1. Eln, Tnc, and Nid2 were abundant in the female OF, while Lama2, Lama5, Lamb2, and Lamc1 were more abundant in the male IF. The complex protein signature of the OF suggests areas of tissue stress, stiffness, and regulatory proteins for growth factor signaling. The higher concentrations of Col4a1 and Col4a2 and their role in steroidogenesis should be further investigated in IF. These findings could significantly enhance our understanding of adrenal cortex functionality and its implications for human health and disease. Key findings were validated, and data are available in ProteomeXchange (PXD046828).
Collapse
Affiliation(s)
- Jean Lucas Kremer
- Institute of Biomedical Sciences, Department of Anatomy, University of São Paulo, Av. Prof. Lineu Prestes, 2415, Butantã, São Paulo, SP 05508-000, Brazil
| | - Veronica Feijoli Santiago
- Institute of Biomedical Sciences, Department of Parasitology, University of São Paulo, Av. Prof. Lineu Prestes, 1374, Butantã, São Paulo, SP 05508-000, Brazil
| | - Fernanda Bongiovani Rodrigues
- Institute of Biomedical Sciences, Department of Anatomy, University of São Paulo, Av. Prof. Lineu Prestes, 2415, Butantã, São Paulo, SP 05508-000, Brazil
| | - Thais Barabba Auricino
- Institute of Biomedical Sciences, Department of Anatomy, University of São Paulo, Av. Prof. Lineu Prestes, 2415, Butantã, São Paulo, SP 05508-000, Brazil
| | - Danilo Henriques de Oliveira Freitas
- Institute of Biomedical Sciences, Department of Anatomy, University of São Paulo, Av. Prof. Lineu Prestes, 2415, Butantã, São Paulo, SP 05508-000, Brazil
| | - Giuseppe Palmisano
- Institute of Biomedical Sciences, Department of Parasitology, University of São Paulo, Av. Prof. Lineu Prestes, 1374, Butantã, São Paulo, SP 05508-000, Brazil
| | - Claudimara Ferini Pacicco Lotfi
- Institute of Biomedical Sciences, Department of Anatomy, University of São Paulo, Av. Prof. Lineu Prestes, 2415, Butantã, São Paulo, SP 05508-000, Brazil
| |
Collapse
|
5
|
Peixoto DO, Bittencourt RR, Gasparotto J, Kessler FGC, Brum PO, Somensi N, Girardi CS, Dos Santos da Silva L, Outeiro TF, Moreira JCF, Gelain DP. Increased alpha-synuclein and neuroinflammation in the substantia nigra triggered by systemic inflammation are reversed by targeted inhibition of the receptor for advanced glycation end products (RAGE). J Neurochem 2024; 168:1587-1607. [PMID: 37661637 DOI: 10.1111/jnc.15956] [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: 03/16/2023] [Revised: 07/18/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023]
Abstract
The receptor for advanced glycation end products (RAGE) is a protein of the immunoglobulin superfamily capable of regulating inflammation. Considering the role of this receptor in the initiation and establishment of neuroinflammation, and the limited understanding of the function of RAGE in the maintenance of this condition, this study describes the effects of RAGE inhibition in the brain, through an intranasal treatment with the antagonist FPS-ZM1, in an animal model of chronic neuroinflammation induced by acute intraperitoneal injection of lipopolysaccharide (LPS). Seventy days after LPS administration (2 mg/kg, i.p.), Wistar rats received, intranasally, 1.2 mg of FPS-ZM1 over 14 days. On days 88 and 89, the animals were submitted to the open-field test and were killed on day 90 after the intraperitoneal injection of LPS. Our results indicate that blockade of encephalic RAGE attenuates LPS-induced chronic neuroinflammation in different brain regions. Furthermore, we found that intranasal FPS-ZM1 administration reduced levels of gliosis markers, RAGE ligands, and α-synuclein in the substantia nigra pars compacta. Additionally, the treatment also reversed the increase in S100 calcium-binding protein B (RAGE ligand) in the cerebrospinal fluid and the cognitive-behavioral deficits promoted by LPS-less time spent in the central zone of the open-field arena (more time in the lateral zones), decreased total distance traveled, and increased number of freezing episodes. In summary, our study demonstrates the prominent role of RAGE in the maintenance of a chronic neuroinflammatory state triggered by a single episode of systemic inflammation and also points to possible future RAGE-based therapeutic approaches to treat conditions in which chronic neuroinflammation and increased α-synuclein levels could play a relevant role, such as in Parkinson's disease.
Collapse
Affiliation(s)
- Daniel Oppermann Peixoto
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Alicante, Spain
| | - Reykla Ramon Bittencourt
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Juciano Gasparotto
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (ICB-UNIFAL), Alfenas, Brazil
| | - Flávio Gabriel Carazza Kessler
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | | | - Nauana Somensi
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Carolina Saibro Girardi
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Lucas Dos Santos da Silva
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Natural Sciences, Göttingen, Germany
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
- Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - José Cláudio Fonseca Moreira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Daniel Pens Gelain
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| |
Collapse
|
6
|
Graves LE, van Dijk EB, Zhu E, Koyyalamudi S, Wotton T, Sung D, Srinivasan S, Ginn SL, Alexander IE. AAV-delivered hepato-adrenal cooperativity in steroidogenesis: Implications for gene therapy for congenital adrenal hyperplasia. Mol Ther Methods Clin Dev 2024; 32:101232. [PMID: 38558568 PMCID: PMC10979120 DOI: 10.1016/j.omtm.2024.101232] [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: 10/13/2023] [Accepted: 03/08/2024] [Indexed: 04/04/2024]
Abstract
Despite the availability of life-saving corticosteroids for 70 years, treatment for adrenal insufficiency is not able to recapitulate physiological diurnal cortisol secretion and results in numerous complications. Gene therapy is an attractive possibility for monogenic adrenocortical disorders such as congenital adrenal hyperplasia; however, requires further development of gene transfer/editing technologies and knowledge of the target progenitor cell populations. Vectors based on adeno-associated virus are the leading system for direct in vivo gene delivery but have limitations in targeting replicating cell populations such as in the adrenal cortex. One strategy to overcome this technological limitation is to deliver the relevant adrenocortical gene to a currently targetable organ outside of the adrenal cortex. To explore this possibility, we developed a vector encoding human 21-hydroxylase and directed expression to the liver in a mouse model of congenital adrenal hyperplasia. This extra-adrenal expression resulted in reconstitution of the steroidogenic pathway. Aldosterone and renin levels normalized, and corticosterone levels improved sufficiently to reduce adrenal hyperplasia. This strategy could provide an alternative treatment option for monogenic adrenal disorders, particularly for mineralocorticoid defects. These findings also demonstrate, when targeting the adrenal gland, that inadvertent liver transduction should be precluded as it may confound data interpretation.
Collapse
Affiliation(s)
- Lara E. Graves
- Gene Therapy Research Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children’s Hospitals Network, Westmead, NSW 2145, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
- Institute of Endocrinology and Diabetes, The Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
| | - Eva B. van Dijk
- Gene Therapy Research Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children’s Hospitals Network, Westmead, NSW 2145, Australia
| | - Erhua Zhu
- Gene Therapy Research Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children’s Hospitals Network, Westmead, NSW 2145, Australia
| | - Sundar Koyyalamudi
- Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
- Institute of Endocrinology and Diabetes, The Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
| | - Tiffany Wotton
- NSW Newborn Screening Program, The Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
| | - Dinah Sung
- NSW Newborn Screening Program, The Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
| | - Shubha Srinivasan
- Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
- Institute of Endocrinology and Diabetes, The Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
| | - Samantha L. Ginn
- Gene Therapy Research Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children’s Hospitals Network, Westmead, NSW 2145, Australia
| | - Ian E. Alexander
- Gene Therapy Research Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children’s Hospitals Network, Westmead, NSW 2145, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| |
Collapse
|
7
|
Augsburger P, Liimatta J, Flück CE. Update on Adrenarche-Still a Mystery. J Clin Endocrinol Metab 2024; 109:1403-1422. [PMID: 38181424 DOI: 10.1210/clinem/dgae008] [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: 09/11/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 01/07/2024]
Abstract
CONTEXT Adrenarche marks the timepoint of human adrenal development when the cortex starts secreting androgens in increasing amounts, in healthy children at age 8-9 years, with premature adrenarche (PA) earlier. Because the molecular regulation and significance of adrenarche are unknown, this prepubertal event is characterized descriptively, and PA is a diagnosis by exclusion with unclear long-term consequences. EVIDENCE ACQUISITION We searched the literature of the past 5 years, including original articles, reviews, and meta-analyses from PubMed, ScienceDirect, Web of Science, Embase, and Scopus, using search terms adrenarche, pubarche, DHEAS, steroidogenesis, adrenal, and zona reticularis. EVIDENCE SYNTHESIS Numerous studies addressed different topics of adrenarche and PA. Although basic studies on human adrenal development, zonation, and zona reticularis function enhanced our knowledge, the exact mechanism leading to adrenarche remains unsolved. Many regulators seem involved. A promising marker of adrenarche (11-ketotestosterone) was found in the 11-oxy androgen pathway. By current definition, the prevalence of PA can be as high as 9% to 23% in girls and 2% to 10% in boys, but only a subset of these children might face related adverse health outcomes. CONCLUSION New criteria for defining adrenarche and PA are needed to identify children at risk for later disease and to spare children with a normal variation. Further research is therefore required to understand adrenarche. Prospective, long-term studies should characterize prenatal or early postnatal developmental pathways that modulate trajectories of birth size, early postnatal growth, childhood overweight/obesity, adrenarche and puberty onset, and lead to abnormal sexual maturation, fertility, and other adverse outcomes.
Collapse
Affiliation(s)
- Philipp Augsburger
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Jani Liimatta
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland and Kuopio University Hospital, 70029 Kuopio, Finland
| | - Christa E Flück
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
8
|
Zucão MI, Grigio V, Guerra LHA, Antoniassi JQ, Castro NFDC, Taboga SR, Vilamaior PSL. Aging effects in adrenal cortex of male Mongolian gerbil: A model for endocrine studies. Steroids 2024; 203:109366. [PMID: 38242273 DOI: 10.1016/j.steroids.2024.109366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
The adrenal gland produces steroid hormones that act in the homeostasis of organisms. During aging, alterations in the hormonal balance affect the adrenal glands, but these have not yet been fully described due to the lack of adequate animal models. The adrenal gland of the Mongolian gerbil has a morphology similar to the primate's adrenal gland, which makes it a possible animal model for endocrine studies. Therefore, the current study aimed to study the morphophysiology of the adrenal gland under the effect of aging. For this purpose, males Meriones unguiculatus, aged three, six, nine, twelve, and fifteen months were used. Morphometric, immunohistochemical, and hormonal analyses were performed. It was observed that during aging the adrenal gland presents hypertrophy of the fasciculata and reticularis zones. Lipofuscin accumulation was observed during aging, in addition to changes in proliferation, cell death, and cell receptors. The analyses also showed that the gerbil presents steroidogenic enzymes and the production of steroid hormones, such as DHEA, like that found in humans. The data provide the first comprehensive assessment of the morphophysiology of the Mongolian gerbil adrenal cortex during aging, indicating that this species is a possible experimental model for studies of the adrenal gland and aging.
Collapse
Affiliation(s)
- Mariele Ilario Zucão
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São Paulo, Brazil
| | - Vitor Grigio
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São Paulo, Brazil
| | - Luiz Henrique Alves Guerra
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São Paulo, Brazil
| | - Julia Quilles Antoniassi
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), São Paulo, Brazil
| | - Nayara Fernanda da Costa Castro
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São Paulo, Brazil
| | - Sebastião Roberto Taboga
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São Paulo, Brazil; Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), São Paulo, Brazil
| | - Patricia Simone Leite Vilamaior
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São Paulo, Brazil.
| |
Collapse
|
9
|
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: 9] [Impact Index Per Article: 4.5] [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
|
10
|
Graves LE, Torpy DJ, Coates PT, Alexander IE, Bornstein SR, Clarke B. Future directions for adrenal insufficiency: cellular transplantation and genetic therapies. J Clin Endocrinol Metab 2023; 108:1273-1289. [PMID: 36611246 DOI: 10.1210/clinem/dgac751] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 01/09/2023]
Abstract
Primary adrenal insufficiency occurs in 1 in 5-7000 adults. Leading aetiologies are autoimmune adrenalitis in adults and congenital adrenal hyperplasia (CAH) in children. Oral replacement of cortisol is lifesaving, but poor quality of life, repeated adrenal crises and dosing uncertainty related to lack of a validated biomarker for glucocorticoid sufficiency, persists. Adrenocortical cell therapy and gene therapy may obviate many of the shortcomings of adrenal hormone replacement. Physiological cortisol secretion regulated by pituitary adrenocorticotropin, could be achieved through allogeneic adrenocortical cell transplantation, production of adrenal-like steroidogenic cells from either stem cells or lineage conversion of differentiated cells, or for CAH, gene therapy to replace or repair a defective gene. The adrenal cortex is a high turnover organ and thus failure to incorporate progenitor cells within a transplant will ultimately result in graft exhaustion. Identification of adrenocortical progenitor cells is equally important in gene therapy where new genetic material must be specifically integrated into the genome of progenitors to ensure a durable effect. Delivery of gene editing machinery and a donor template, allowing targeted correction of the 21-hydroxylase gene, has the potential to achieve this. This review describes advances in adrenal cell transplants and gene therapy that may allow physiological cortisol production for children and adults with primary adrenal insufficiency.
Collapse
Affiliation(s)
- Lara E Graves
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, NSW, Australia
- Gene Therapy Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children's Hospitals Network, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - David J Torpy
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - P Toby Coates
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Central Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Ian E Alexander
- Gene Therapy Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children's Hospitals Network, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Stefan R Bornstein
- University Clinic Carl Gustav Carus, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Brigette Clarke
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
11
|
Lerario AM, Mohan DR, Hammer GD. Update on Biology and Genomics of Adrenocortical Carcinomas: Rationale for Emerging Therapies. Endocr Rev 2022; 43:1051-1073. [PMID: 35551369 PMCID: PMC9695111 DOI: 10.1210/endrev/bnac012] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Indexed: 11/19/2022]
Abstract
The adrenal glands are paired endocrine organs that produce steroid hormones and catecholamines required for life. Adrenocortical carcinoma (ACC) is a rare and often fatal cancer of the peripheral domain of the gland, the adrenal cortex. Recent research in adrenal development, homeostasis, and disease have refined our understanding of the cellular and molecular programs controlling cortical growth and renewal, uncovering crucial clues into how physiologic programs are hijacked in early and late stages of malignant neoplasia. Alongside these studies, genome-wide approaches to examine adrenocortical tumors have transformed our understanding of ACC biology, and revealed that ACC is composed of distinct molecular subtypes associated with favorable, intermediate, and dismal clinical outcomes. The homogeneous transcriptional and epigenetic programs prevailing in each ACC subtype suggest likely susceptibility to any of a plethora of existing and novel targeted agents, with the caveat that therapeutic response may ultimately be limited by cancer cell plasticity. Despite enormous biomedical research advances in the last decade, the only potentially curative therapy for ACC to date is primary surgical resection, and up to 75% of patients will develop metastatic disease refractory to standard-of-care adjuvant mitotane and cytotoxic chemotherapy. A comprehensive, integrated, and current bench-to-bedside understanding of our field's investigations into adrenocortical physiology and neoplasia is crucial to developing novel clinical tools and approaches to equip the one-in-a-million patient fighting this devastating disease.
Collapse
Affiliation(s)
- Antonio Marcondes Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | - Dipika R Mohan
- Medical Scientist Training Program, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| |
Collapse
|
12
|
Zhang K, Hu Y, Li R, Li T. Single-cell atlas of murine adrenal glands reveals immune-adrenal crosstalk during systemic <i>Candida albicans</i> infection. Front Immunol 2022; 13:966814. [PMID: 36389688 PMCID: PMC9664004 DOI: 10.3389/fimmu.2022.966814] [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: 06/11/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
Fungal sepsis remains a major health threat with high mortality, where the adrenal gland stress response has been rarely reported. <i>Candida albicans</i> (<i>C.albicans</i>) is the most common opportunistic fungal pathogen of life-threatening disseminated candidiasis and fungal sepsis. In the present study, we performed single-cell RNA sequencing (scRNA-Seq) using the 10x Genomics platform to analyze the changes in murine adrenal transcriptome following systemic <i>C.albicans</i> infection. A total of 16 021 cells were categorized into 18 transcriptionally distinct clusters, representing adrenocortical cells, endothelial cells, various immune cells, mesenchymal cells, smooth muscle cells, adrenal capsule, chromaffin cells, neurons and glials. As the main cell component in the adrenal gland responsible for steroidogenesis, the adrenocortical cells dramatically diminished and were further grouped into 10 subclusters, which differently distributed in the infected and uninfected samples. Pseudo-time analysis revealed transitions of the adrenocortical cells from the initial normal states to active or dysfunctional states following systemic <i>C.albicans</i> infection <i>via</i> two trajectory paths. Endothelial cells in the highly vascularized organ of adrenal gland further proliferated following infection, with the upregulation of genes positively regulating angiogenesis and downregulation of protective genes of endothelial cells. Immune cells were also excessively infiltrated in adrenal glands of <i>C.albicans</i>-infected mice. Macrophages dominated the immune microenvironments in murine adrenal glands both before and after <i>C.albicans</i> infection, mediating the crosstalk among the steroid-producing cells, endothelial cells and immune cells within the adrenal gland. NLR family, pyrin domain containing 3 (NLRP3, encoded by <i>Nlrp3</i>) and complement receptor 3 (CR3, encoded by <i>Itgam</i>) were found to be significantly upregulated on the adrenal macrophages upon systemic <i>C.albicans</i> infection and might play critical roles in mediating the myeloid response. Meanwhile, the number and strength of the interactions between the infiltrating immune cells and adrenal resident cells were unveiled by cell-cell communication analysis to be dramatically increased after systemic <i>C.albicans</i> infection, indicating that the immune-adrenal crosstalk might contribute to the compromised functions of adrenal cells. Overall, our comprehensive picture of the murine adrenal gland microenvironment in systemic <i>C.albicans</i> infection provides deeper insights into the immune-adrenal cell communications during fungal sepsis.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China,National Clinical Research Center for Skin and Immune Diseases, Beijing, China,Research Center for Medical Mycology, Peking University, Beijing, China,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China
| | - Yuzhe Hu
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China,Key Laboratory of Medical Immunology, National Health Commission of the People's Republic of China, Beijing, China,Peking University Center for Human Disease Genomics, Beijing, China
| | - Ruoyu Li
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China,National Clinical Research Center for Skin and Immune Diseases, Beijing, China,Research Center for Medical Mycology, Peking University, Beijing, China,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China,*Correspondence: Ting Li, ; Ruoyu Li,
| | - Ting Li
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China,Key Laboratory of Medical Immunology, National Health Commission of the People's Republic of China, Beijing, China,Peking University Center for Human Disease Genomics, Beijing, China,*Correspondence: Ting Li, ; Ruoyu Li,
| |
Collapse
|
13
|
Abstract
Resident progenitor and/or stem cell populations in the adult adrenal cortex enable cortical cells to undergo homeostatic renewal and regeneration after injury. Renewal occurs predominantly in the outer layers of the adrenal gland but newly formed cells undergo centripetal migration, differentiation and lineage conversion in the process of forming the different functional steroidogenic zones. Over the past 10 years, advances in the genetic characterization of adrenal diseases and studies of mouse models with altered adrenal phenotypes have helped to elucidate the molecular pathways that regulate adrenal tissue renewal, several of which are fine-tuned via complex paracrine and endocrine influences. Moreover, the adrenal gland is a sexually dimorphic organ, and testicular androgens have inhibitory effects on cell proliferation and progenitor cell recruitment in the adrenal cortex. This Review integrates these advances, including the emerging role of sex hormones, into existing knowledge on adrenocortical cell renewal. An in-depth understanding of these mechanisms is expected to contribute to the development of novel therapies for severe endocrine diseases, for which current treatments are unsatisfactory.
Collapse
Affiliation(s)
- Rodanthi Lyraki
- Université Côte d'Azur, INSERM, CNRS, Institut de Biologie Valrose, Nice, France
| | - Andreas Schedl
- Université Côte d'Azur, INSERM, CNRS, Institut de Biologie Valrose, Nice, France.
| |
Collapse
|
14
|
Beta-Catenin Causes Adrenal Hyperplasia by Blocking Zonal Transdifferentiation. Cell Rep 2021; 31:107524. [PMID: 32320669 PMCID: PMC7281829 DOI: 10.1016/j.celrep.2020.107524] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/16/2020] [Accepted: 03/26/2020] [Indexed: 12/19/2022] Open
Abstract
Activating mutations in the canonical Wnt/β-catenin pathway are key drivers of hyperplasia, the gateway for tumor development. In a wide range of tissues, this occurs primarily through enhanced effects on cellular proliferation. Whether additional mechanisms contribute to β-catenin-driven hyperplasia remains unknown. The adrenal cortex is an ideal system in which to explore this question, as it undergoes hyperplasia following somatic β-catenin gain-of-function (βcat-GOF) mutations. Targeting βcat-GOF to zona Glomerulosa (zG) cells leads to a progressive hyperplastic expansion in the absence of increased proliferation. Instead, we find that hyperplasia results from a functional block in the ability of zG cells to transdifferentiate into zona Fasciculata (zF) cells. Mechanistically, zG cells demonstrate an upregulation of Pde2a, an inhibitor of zF-specific cAMP/PKA signaling. Hyperplasia is further exacerbated by trophic factor stimulation leading to organomegaly. Together, these data indicate that β-catenin drives adrenal hyperplasia through both proliferation-dependent and -independent mechanisms. Using the adrenal cortex as a model for slow-cycling tissues, Pignatti et al. show that activation of the canonical Wnt/β-catenin pathway leads to tissue hyperplasia by blocking cellular differentiation/cell-fate commitment, independent of its effects on cellular proliferation.
Collapse
|
15
|
The Sexually Dimorphic Adrenal Cortex: Implications for Adrenal Disease. Int J Mol Sci 2021; 22:ijms22094889. [PMID: 34063067 PMCID: PMC8124132 DOI: 10.3390/ijms22094889] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Many adrenocortical diseases are more prevalent in women than in men, but the reasons underlying this sex bias are still unknown. Recent studies involving gonadectomy and sex hormone replacement experiments in mice have shed some light onto the molecular basis of sexual dimorphism in the adrenal cortex. Indeed, it has been shown that gonadal hormones influence many aspects of adrenal physiology, ranging from stem cell-dependent tissue turnover to steroidogenesis and X-zone dynamics. This article reviews current knowledge on adrenal cortex sexual dimorphism and the potential mechanisms underlying sex hormone influence of adrenal homeostasis. Both topics are expected to contribute to personalized and novel therapeutic approaches in the future.
Collapse
|
16
|
Sbiera I, Kircher S, Altieri B, Fassnacht M, Kroiss M, Sbiera S. Epithelial and Mesenchymal Markers in Adrenocortical Tissues: How Mesenchymal Are Adrenocortical Tissues? Cancers (Basel) 2021; 13:1736. [PMID: 33917436 PMCID: PMC8038668 DOI: 10.3390/cancers13071736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/02/2021] [Accepted: 04/04/2021] [Indexed: 12/22/2022] Open
Abstract
A clinically relevant proportion of adrenocortical carcinoma (ACC) cases shows a tendency to metastatic spread. The objective was to determine whether the epithelial to mesenchymal transition (EMT), a mechanism associated with metastasizing in several epithelial cancers, might play a crucial role in ACC. 138 ACC, 29 adrenocortical adenomas (ACA), three normal adrenal glands (NAG), and control tissue samples were assessed for the expression of epithelial (E-cadherin and EpCAM) and mesenchymal (N-cadherin, SLUG and SNAIL) markers by immunohistochemistry. Using real-time RT-PCR we quantified the alternative isoform splicing of FGFR 2 and 3, another known indicator of EMT. We also assessed the impact of these markers on clinical outcome. Results show that both normal and neoplastic adrenocortical tissues lacked expression of epithelial markers but strongly expressed mesenchymal markers N-cadherin and SLUG. FGFR isoform splicing confirmed higher similarity of adrenocortical tissues to mesenchymal compared to epithelial tissues. In ACC, higher SLUG expression was associated with clinical markers indicating aggressiveness, while N-cadherin expression inversely associated with these markers. In conclusion, we could not find any indication of EMT as all adrenocortical tissues lacked expression of epithelial markers and exhibited closer similarity to mesenchymal tissues. However, while N-cadherin might play a positive role in tissue structure upkeep, SLUG seems to be associated with a more aggressive phenotype.
Collapse
Affiliation(s)
- Iuliu Sbiera
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, 97080 Würzburg, Germany; (I.S.); (B.A.); (M.F.)
| | - Stefan Kircher
- Institute for Pathology, University of Würzburg, 97080 Würzburg, Germany;
| | - Barbara Altieri
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, 97080 Würzburg, Germany; (I.S.); (B.A.); (M.F.)
| | - Martin Fassnacht
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, 97080 Würzburg, Germany; (I.S.); (B.A.); (M.F.)
- Clinical Chemistry and Laboratory Medicine, University Hospital Würzburg, 97080 Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Würzburg, 97080 Würzburg, Germany
| | - Matthias Kroiss
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, 97080 Würzburg, Germany; (I.S.); (B.A.); (M.F.)
- Comprehensive Cancer Center Mainfranken, University of Würzburg, 97080 Würzburg, Germany
- Department of Internal Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Silviu Sbiera
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital Würzburg, 97080 Würzburg, Germany; (I.S.); (B.A.); (M.F.)
| |
Collapse
|
17
|
Oikonomakos I, Weerasinghe Arachchige LC, Schedl A. Developmental mechanisms of adrenal cortex formation and their links with adult progenitor populations. Mol Cell Endocrinol 2021; 524:111172. [PMID: 33484742 DOI: 10.1016/j.mce.2021.111172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/15/2020] [Accepted: 01/13/2021] [Indexed: 12/16/2022]
Abstract
The adrenal cortex is the main steroid producing organ of the human body. Studies on adrenal tissue renewal have been neglected for many years, but recent intensified research has seen tremendous progress in our understanding of the formation and homeostasis of this organ. However, cell turnover of the adrenal cortex appears to be complex and several cell populations have been identified that can differentiate into steroidogenic cells and contribute to adrenal cortex renewal. The purpose of this review is to provide an overview of how the adrenal cortex develops and how stem cell populations relate to its developmental progenitors. Finally, we will summarize present and future approaches to harvest the potential of progenitor/stem cells for future cell replacement therapies.
Collapse
Affiliation(s)
- Ioannis Oikonomakos
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, 06108, Nice, France.
| | | | - Andreas Schedl
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, 06108, Nice, France.
| |
Collapse
|
18
|
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
|
19
|
Gannon AL, O’Hara L, Mason IJ, Jørgensen A, Frederiksen H, Curley M, Milne L, Smith S, Mitchell RT, Smith LB. Androgen Receptor Is Dispensable for X-Zone Regression in the Female Adrenal but Regulates Post-Partum Corticosterone Levels and Protects Cortex Integrity. Front Endocrinol (Lausanne) 2021; 11:599869. [PMID: 33584538 PMCID: PMC7873917 DOI: 10.3389/fendo.2020.599869] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/24/2020] [Indexed: 01/11/2023] Open
Abstract
Adrenal androgens are fundamental mediators of ovarian folliculogenesis, embryonic implantation, and breast development. Although adrenal androgen function in target tissues are well characterized, there is little research covering the role of androgen-signaling within the adrenal itself. Adrenal glands express AR which is essential for the regression of the X-zone in male mice. Female mice also undergo X-zone regression during their first pregnancy, however whether this is also controlled by AR signaling is unknown. To understand the role of the androgen receptor (AR) in the female adrenal, we utilized a Cyp11a1-Cre to specifically ablate AR from the mouse adrenal cortex. Results show that AR-signaling is dispensable for adrenal gland development in females, and for X-zone regression during pregnancy, but is required to suppress elevation of corticosterone levels post-partum. Additionally, following disruption to adrenal AR, aberrant spindle cell development is observed in young adult females. These results demonstrate sexually dimorphic regulation of the adrenal X-zone by AR and point to dysfunctional adrenal androgen signaling as a possible mechanism in the early development of adrenal spindle cell hyperplasia.
Collapse
Affiliation(s)
- Anne-Louise Gannon
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
- School of Environmental and Life Sciences, Faculty of Science, University of Newcastle, Callaghan, NSW, Australia
| | - Laura O’Hara
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Hugh Robson Building, George Square, Edinburgh, United Kingdom
| | - Ian J. Mason
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Anne Jørgensen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, Copenhagen, Denmark
| | - Hanne Frederiksen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, Copenhagen, Denmark
| | - Michael Curley
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Laura Milne
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Sarah Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Rod T. Mitchell
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Lee B. Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
- School of Environmental and Life Sciences, Faculty of Science, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
20
|
Taylor MJ, Ullenbruch MR, Frucci EC, Rege J, Ansorge MS, Gomez-Sanchez CE, Begum S, Laufer E, Breault DT, Rainey WE. Chemogenetic activation of adrenocortical Gq signaling causes hyperaldosteronism and disrupts functional zonation. J Clin Invest 2020; 130:83-93. [PMID: 31738186 DOI: 10.1172/jci127429] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 09/18/2019] [Indexed: 02/04/2023] Open
Abstract
The mineralocorticoid aldosterone is produced in the adrenal zona glomerulosa (ZG) under the control of the renin-angiotensin II (AngII) system. Primary aldosteronism (PA) results from renin-independent production of aldosterone and is a common cause of hypertension. PA is caused by dysregulated localization of the enzyme aldosterone synthase (Cyp11b2), which is normally restricted to the ZG. Cyp11b2 transcription and aldosterone production are predominantly regulated by AngII activation of the Gq signaling pathway. Here, we report the generation of transgenic mice with Gq-coupled designer receptors exclusively activated by designer drugs (DREADDs) specifically in the adrenal cortex. We show that adrenal-wide ligand activation of Gq DREADD receptors triggered disorganization of adrenal functional zonation, with induction of Cyp11b2 in glucocorticoid-producing zona fasciculata cells. This result was consistent with increased renin-independent aldosterone production and hypertension. All parameters were reversible following termination of DREADD-mediated Gq signaling. These findings demonstrate that Gq signaling is sufficient for adrenocortical aldosterone production and implicate this pathway in the determination of zone-specific steroid production within the adrenal cortex. This transgenic mouse also provides an inducible and reversible model of hyperaldosteronism to investigate PA therapeutics and the mechanisms leading to the damaging effects of aldosterone on the cardiovascular system.
Collapse
Affiliation(s)
- Matthew J Taylor
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew R Ullenbruch
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Emily C Frucci
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Juilee Rege
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Mark S Ansorge
- The Sackler Institute for Developmental Psychobiology, Columbia University, New York, New York, USA
| | - Celso E Gomez-Sanchez
- Endocrine Section, G.V. (Sonny) Montgomery VA Medical Center and the Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Salma Begum
- Department of Obstetrics, Gynecology and Women's Health, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Edward Laufer
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | - David T Breault
- Department of Pediatrics, Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - William E Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA.,Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
21
|
Gallol LE, Busolini FI, Mohamed FH. Influence of melatonin and sexual hormones on the expression of proliferating cell nuclear antigen in the adrenal cortex of a seasonal breeder (Lagostomus maximus). Anat Rec (Hoboken) 2020; 303:3052-3067. [PMID: 32445505 DOI: 10.1002/ar.24457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/25/2020] [Accepted: 03/31/2020] [Indexed: 12/14/2022]
Abstract
The viscacha (Lagostomus maximus) is a rodent of nocturnal habits, whose physiology and behavior vary according to modifications of environmental signals. The objective of this study is to assess the influence of melatonin and sexual hormones on the viscacha adrenal cortex proliferative activity through the immunohistochemical detection of proliferating cell nuclear antigen (PCNA) along with hormonal determinations. PCNA expression was studied in male viscachas to assess the effect of melatonin administration, castration, and the annual reproductive cycle. In female viscachas, PCNA was studied in nonpregnant and pregnant viscachas. PCNA expression was observed in adrenocortical cells (PCNA-A) and endothelial cells (PCNA-E). Melatonin-administered animals showed a significantly lower number of PCNA-A compared to the control group. No significant difference could be established in the number of PCNA-A and PCNA-E between castrated and control animals. However, the morphometric analysis showed an increase in the size of the cortex of castrated animals, along with other cytological features. Significant differences in serum testosterone levels were observed during the male viscacha reproductive cycle, with the lowest levels encountered during the regression period (winter). Male viscachas exhibited a significantly high number of PCNA-A during late autumn and a high number of PCNA-E during winter. In females, hormonal determinations showed a peak of progesterone and estrogen during mid-pregnancy, along with a notably high number of PCNA-A and an increase in the number of PCNA-E. Our results suggest that proliferation in the adrenal cortex of the viscacha varies in relation to melatonin, sexual hormones, and environmental conditions.
Collapse
Affiliation(s)
- Luis Ezequiel Gallol
- Cátedra de Histología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, 5700, Argentina.,Centro Científico Tecnológico del Consejo Nacional de Investigaciones Científicas y Técnicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis, 5700, Argentina
| | - Fabricio Iván Busolini
- Cátedra de Histología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, 5700, Argentina.,Centro Científico Tecnológico del Consejo Nacional de Investigaciones Científicas y Técnicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis, 5700, Argentina
| | - Fabian Heber Mohamed
- Cátedra de Histología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, 5700, Argentina
| |
Collapse
|
22
|
Wu K, Li Y, Liu J, Mo J, Li X, Ge RS. Long-term triphenyltin exposure disrupts adrenal function in adult male rats. CHEMOSPHERE 2020; 243:125149. [PMID: 31765896 DOI: 10.1016/j.chemosphere.2019.125149] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/30/2019] [Accepted: 10/17/2019] [Indexed: 06/10/2023]
Abstract
Triphenyltin is an organotin, which is widely used as a fungicide in agriculture. Here, we reported the effects of triphenyltin on adrenal function in adult male rats. Adult male Sprague Dawley rats were daily gavaged with triphenyltin (0, 0.5, 1, and 2 mg/kg body weight) from postnatal day 56-86. Triphenyltin significantly decreased serum corticosterone levels at 1 and 2 mg/kg without affecting serum levels of aldosterone and adrenocorticotropic hormone. Triphenyltin increased thickness of zona glomerulosa without affecting that of zona fasciculata. Triphenyltin did not affect cell number in zona fasciculata and zona glomerulosa. Triphenyltin down-regulated the expression of Scarb1, Star, Cyp11a1, Hsd3b1, Cyp21, Cyp11b1, and Hsd11b1 at 1 and/or 2 mg/kg while it up-regulated the expression of At1, Nr4a2, and Hsd11b2 at 2 mg/kg. Triphenyltin activated the phosphorylation of AMPKα while suppressed the phosphorylation of AKT1 and SIRT1/PGC-1α in rat adrenals in vivo and H295R cells in vitro. In vitro, triphenyltin also induced ROS production in H295R cells at 100 nM, a concentration at which no apoptosis was induced. In conclusion, triphenyltin disrupts glucocorticoid synthesis in rat adrenal cortex via several mechanisms: 1) lowering AKT1 phosphorylation and SIRT1/PGC-1α levels; 2) activating AMPKα; and 3) possibly inducing ROS production.
Collapse
Affiliation(s)
- Keyang Wu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yang Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jianpeng Liu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jiaying Mo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Xiaoheng Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Ren-Shan Ge
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
23
|
Gannon AL, O'Hara L, Mason JI, Jørgensen A, Frederiksen H, Milne L, Smith S, Mitchell RT, Smith LB. Androgen receptor signalling in the male adrenal facilitates X-zone regression, cell turnover and protects against adrenal degeneration during ageing. Sci Rep 2019; 9:10457. [PMID: 31320667 PMCID: PMC6639311 DOI: 10.1038/s41598-019-46049-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 06/20/2019] [Indexed: 11/09/2022] Open
Abstract
Androgens are known to be an essential regulator of male health. Androgen receptor (AR) is widely expressed throughout the adrenal cortex, yet the wider role for androgen signalling in the adrenal remains underexplored. To investigate AR-dependent and AR-independent androgen signalling in the adrenal, we used a novel mouse model with a specific ablation of androgen receptor in the adrenal cortex with or without reduction of circulating androgen levels by castration. Our results describe AR expression in the human and mouse adrenal and highlight that the mouse is a viable model to investigate androgen signalling in the adrenal cortex. We show androgen signalling via AR is required for X-zone regression during puberty. Furthermore, cortex measurements define differences in X-zone morphology depending on whether circulating androgens or AR have been removed. We show androgens promote both cortical cell differentiation and apoptosis but are dispensable for the formation of the definitive cortex. Additionally, investigation of aged mice with AR ablation reveals severe cortex disruption, spindle cell hyperplasia and X-zone expansion. The data described herein demonstrates AR-signalling is required to facilitate X-zone regression, cell clearance and to protect against adrenal degeneration during ageing.
Collapse
Affiliation(s)
- Anne-Louise Gannon
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
- School of Environmental and Life Sciences, Faculty of Science, University of Newcastle, Callaghan, 2308, NSW, Australia
| | - Laura O'Hara
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
- Centre for Discovery Brain Sciences, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - J Ian Mason
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Anne Jørgensen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, Copenhagen, Denmark
| | - Hanne Frederiksen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, Copenhagen, Denmark
| | - Laura Milne
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
- Edinburgh Genome Foundry, Michael Swann Building, Max Bonn Crescent, Edinburgh, EH9 3BF, UK
| | - Sarah Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
- School of Environmental and Life Sciences, Faculty of Science, University of Newcastle, Callaghan, 2308, NSW, Australia.
| |
Collapse
|
24
|
Grabek A, Dolfi B, Klein B, Jian-Motamedi F, Chaboissier MC, Schedl A. The Adult Adrenal Cortex Undergoes Rapid Tissue Renewal in a Sex-Specific Manner. Cell Stem Cell 2019; 25:290-296.e2. [PMID: 31104943 DOI: 10.1016/j.stem.2019.04.012] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/13/2019] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
Abstract
Evolution has resulted in profound differences between males and females that extend to non-reproductive organs and are reflected in the susceptibility and progression of diseases. However, the cellular and molecular basis for these differences remains largely unknown. Here we report that adrenal gland tissue renewal is highly active and sexually dimorphic, with female mice showing a 3-fold higher turnover than males. Moreover, in males, homeostasis relies on proliferation of cells within the steroidogenic zone, but females employ an additional stem and/or progenitor compartment situated in the adrenal capsule. Using lineage tracing, sex reversal models, gonadectomy, and dihydrotestosterone treatments, we further show that sex-specific stem cell activity is driven by male hormones that repress recruitment of Gli1+ stem cells from the capsule and cell proliferation. Taken together, our findings provide a molecular and cellular basis for adrenal sex dimorphism that may contribute to the increased incidence of adrenal diseases in females.
Collapse
Affiliation(s)
| | - Bastien Dolfi
- Université Côte d'Azur, INSERM, CNRS, iBV, Nice, France
| | - Bryan Klein
- Université Côte d'Azur, INSERM, CNRS, iBV, Nice, France
| | | | | | - Andreas Schedl
- Université Côte d'Azur, INSERM, CNRS, iBV, Nice, France; Equipe Labellisée Ligue Contre le Cancer.
| |
Collapse
|
25
|
Isolation and characterization of adrenocortical progenitors involved in the adaptation to stress. Proc Natl Acad Sci U S A 2018; 115:12997-13002. [PMID: 30514817 PMCID: PMC6304967 DOI: 10.1073/pnas.1814072115] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Humans are constantly confronted with multiple stressors, to which the bodily response and adaptation are essential. The adrenal gland plays a major role in the response to physiological challenges. Maintenance of the adrenal is partly accomplished by proliferation and differentiation of adult progenitors and stem cells in the cortex and medulla. In this study, we have isolated and characterized a subpopulation of adrenocortical progenitors, which are interconnected with adrenomedullary stress-dependent progenitors. Under stress, the adrenocortical progenitors are also activated and they mobilize, giving rise to steroidogenic cells. Our findings demonstrate the coordinated action of stress-inducible stem cells to ensure tissue remodeling and cellular and functional adaptation to stress. The adrenal gland is a master regulator of the human body during response to stress. This organ shows constant replacement of senescent cells by newly differentiated cells. A high degree of plasticity is critical to sustain homeostasis under different physiological demands. This is achieved in part through proliferation and differentiation of adult adrenal progenitors. Here, we report the isolation and characterization of a Nestin+ population of adrenocortical progenitors located under the adrenal capsule and scattered throughout the cortex. These cells are interconnected with progenitors in the medulla. In vivo lineage tracing revealed that, under basal conditions, this population is noncommitted and slowly migrates centripetally. Under stress, this migration is greatly enhanced, and the cells differentiate into steroidogenic cells. Nestin+ cells cultured in vitro also show multipotency, as they differentiate into mineralocorticoid and glucocorticoid-producing cells, which can be further influenced by the exposure to Angiotensin II, adrenocorticotropic hormone, and the agonist of luteinizing hormone-releasing hormone, triptorelin. Taken together, Nestin+ cells in the adult adrenal cortex exhibit the features of adrenocortical progenitor cells. Our study provides evidence for a role of Nestin+ cells in organ homeostasis and emphasizes their role under stress. This cell population might be a potential source of cell replacement for the treatment of adrenal insufficiency.
Collapse
|
26
|
Lotfi CFP, Kremer JL, dos Santos Passaia B, Cavalcante IP. The human adrenal cortex: growth control and disorders. Clinics (Sao Paulo) 2018; 73:e473s. [PMID: 30208164 PMCID: PMC6113920 DOI: 10.6061/clinics/2018/e473s] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/26/2018] [Indexed: 12/15/2022] Open
Abstract
This review summarizes key knowledge regarding the development, growth, and growth disorders of the adrenal cortex from a molecular perspective. The adrenal gland consists of two distinct regions: the cortex and the medulla. During embryological development and transition to the adult adrenal gland, the adrenal cortex acquires three different structural and functional zones. Significant progress has been made in understanding the signaling and molecules involved during adrenal cortex zonation. Equally significant is the knowledge obtained regarding the action of peptide factors involved in the maintenance of zonation of the adrenal cortex, such as peptides derived from proopiomelanocortin processing, adrenocorticotropin and N-terminal proopiomelanocortin. Findings regarding the development, maintenance and growth of the adrenal cortex and the molecular factors involved has improved the scientific understanding of disorders that affect adrenal cortex growth. Hypoplasia, hyperplasia and adrenocortical tumors, including adult and pediatric adrenocortical adenomas and carcinomas, are described together with findings regarding molecular and pathway alterations. Comprehensive genomic analyses of adrenocortical tumors have shown gene expression profiles associated with malignancy as well as methylation alterations and the involvement of miRNAs. These findings provide a new perspective on the diagnosis, therapeutic possibilities and prognosis of adrenocortical disorders.
Collapse
Affiliation(s)
- Claudimara Ferini Pacicco Lotfi
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| | - Jean Lucas Kremer
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Barbara dos Santos Passaia
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Isadora Pontes Cavalcante
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
27
|
Candida Barisson Villares Fragoso M, Pontes Cavalcante I, Meneses Ferreira A, Marinho de Paula Mariani B, Ferini Pacicco Lotfi C. Genetics of primary macronodular adrenal hyperplasia. Presse Med 2018; 47:e139-e149. [PMID: 30075949 DOI: 10.1016/j.lpm.2018.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent advances in molecular genetics investigations of primary macronodular adrenal hyperplasia (PMAH) have been providing new insights for the research on this issue. The cAMP-dependent pathway is physiologically triggered by ACTH and its receptor, MC2-R, in adrenocortical cells. Different mechanisms of this cascade may be altered in some functioning adrenal cortical disorders. Activating somatic mutations of the GNAS gene (known as gsp oncogene) which encodes the stimulatory G protein alpha-subunit (Gsα) have been found in a small number of adrenocortical secreting adenomas and rarely in PMAH. Lately, ARMC5 was linked to the cyclic AMP signaling pathway, which could be implicated in all of mechanisms of cortisol-secreting by macronodules adrenal hyperplasia and the molecular defects in: G protein aberrant receptors; MC2R; GNAS; PRKAR1A; PDE11A; PDE8B. Around 50 % of patient's relatives with PMAH and 30 % of apparently sporadic hypercortisolism carried ARMC5 mutations. Therefore, PMAH is genetically determined more frequently than previously believed. This review summarizes the most important molecular mechanisms involved in PMAH.
Collapse
Affiliation(s)
| | - Isadora Pontes Cavalcante
- University of Sao Paulo, Adrenal Unit, Service of Endocrinology and Metabolism, 03178-200 Sao Paulo, Brazil; University of Sao Paulo, Institute of Biomedical Sciences, Department of Anatomy, 03178-200 Sao Paulo, Brazil
| | - Amanda Meneses Ferreira
- University of Sao Paulo, Adrenal Unit, Service of Endocrinology and Metabolism, 03178-200 Sao Paulo, Brazil
| | | | | |
Collapse
|
28
|
Markmann S, De BP, Reid J, Jose CL, Rosenberg JB, Leopold PL, Kaminsky SM, Sondhi D, Pagovich O, Crystal RG. Biology of the Adrenal Gland Cortex Obviates Effective Use of Adeno-Associated Virus Vectors to Treat Hereditary Adrenal Disorders. Hum Gene Ther 2018; 29:403-412. [DOI: 10.1089/hum.2017.203] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Sandra Markmann
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Bishnu P. De
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Jasmine Reid
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Clarisse L. Jose
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | | | - Philip L. Leopold
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Stephen M. Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Odelya Pagovich
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald G. Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
29
|
Balyura M, Gelfgat E, Steenblock C, Androutsellis-Theotokis A, Ruiz-Babot G, Guasti L, Werdermann M, Ludwig B, Bornstein T, Schally AV, Brennand A, Bornstein SR. Expression of progenitor markers is associated with the functionality of a bioartificial adrenal cortex. PLoS One 2018; 13:e0194643. [PMID: 29596439 PMCID: PMC5875767 DOI: 10.1371/journal.pone.0194643] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 03/07/2018] [Indexed: 11/19/2022] Open
Abstract
Encapsulation of primary bovine adrenocortical cells in alginate is an efficacious model of a bioartificial adrenal cortex. Such a bioartificial adrenal cortex can be used for the restoration of lost adrenal function in vivo as well as for in vitro modeling of the adrenal microenvironment and for investigation of cell–cell interactions in the adrenals. The aim of this work was the optimization of a bioartificial adrenal cortex, that is the generation of a highly productive, self-regenerating, long-term functioning and immune tolerant bioartificial organ. To achieve this, it is necessary that adrenocortical stem and progenitor cells are present in the bioartificial gland, as these undifferentiated cells play important roles in the function of the mature gland. Here, we verified the presence of adrenocortical progenitors in cultures of bovine adrenocortical cells, studied the dynamics of their appearance and growth and determined the optimal time point for cell encapsulation. These procedures increased the functional life span and reduced the immunogenicity of the bioartificial adrenal cortex. This model allows the use of the luteinizing hormone-releasing hormone (LHRH) agonist triptorelin, the neuropeptide bombesin, and retinoic acid to alter cell number and the release of cortisol over long periods of time.
Collapse
Affiliation(s)
- Mariya Balyura
- University Hospital Carl Gustav Carus, Dept. of Medicine III, Technische Universität Dresden, Dresden, Germany
- * E-mail:
| | - Evgeny Gelfgat
- University Hospital Carl Gustav Carus, Dept. of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Charlotte Steenblock
- University Hospital Carl Gustav Carus, Dept. of Medicine III, Technische Universität Dresden, Dresden, Germany
| | | | - Gerard Ruiz-Babot
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Martin Werdermann
- University Hospital Carl Gustav Carus, Dept. of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Barbara Ludwig
- University Hospital Carl Gustav Carus, Dept. of Medicine III, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of TU Dresden Faculty of Medicine, Dresden, Germany
- Center for Regenerative Therapies, Technische Universität Dresden, Dresden, Germany
| | - Tobias Bornstein
- University Hospital Carl Gustav Carus, Dept. of Medicine III, Technische Universität Dresden, Dresden, Germany
- Diabetes and Nutritional Sciences Division, King's College London, London, United Kingdom
| | - Andrew V. Schally
- Divisions of Endocrinology and Hematology–Oncology, Departments of Medicine and Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL, United States of America
- Veterans Affairs Medical Center, Miami, FL, United States of America
| | - Ana Brennand
- University Hospital Carl Gustav Carus, Dept. of Medicine III, Technische Universität Dresden, Dresden, Germany
- Diabetes and Nutritional Sciences Division, King's College London, London, United Kingdom
| | - Stefan R. Bornstein
- University Hospital Carl Gustav Carus, Dept. of Medicine III, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of TU Dresden Faculty of Medicine, Dresden, Germany
- Center for Regenerative Therapies, Technische Universität Dresden, Dresden, Germany
- Diabetes and Nutritional Sciences Division, King's College London, London, United Kingdom
| |
Collapse
|
30
|
Finco I, Lerario AM, Hammer GD. Sonic Hedgehog and WNT Signaling Promote Adrenal Gland Regeneration in Male Mice. Endocrinology 2018; 159:579-596. [PMID: 29211850 PMCID: PMC5774245 DOI: 10.1210/en.2017-03061] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 11/27/2017] [Indexed: 01/08/2023]
Abstract
The atrophy and hypofunction of the adrenal cortex following long-term pharmacologic glucocorticoid therapy is a major health problem necessitating chronic glucocorticoid replacement that often prolongs the ultimate return of endogenous adrenocortical function. Underlying this functional recovery is anatomic regeneration, the cellular and molecular mechanisms of which are poorly understood. Investigating the lineage contribution of cortical Sonic hedgehog (Shh)+ progenitor cells and the SHH-responsive capsular Gli1+ cells to the regenerating adrenal cortex, we observed a spatially and temporally bimodal contribution of both cell types to adrenocortical regeneration following cessation of glucocorticoid treatment. First, an early repopulation of the cortex is defined by a marked delamination and expansion of capsular Gli1+ cells, recapitulating the establishment of the capsular-cortical homeostatic niche during embryonic development. This rapid repopulation is promptly cleared from the cortical compartment only to be supplanted by repopulating cortical cells derived from the resident long-term-retained zona glomerulosa Shh+ progenitors. Pharmacologic and genetic dissection of SHH signaling further defines an SHH-dependent activation of WNT signaling that supports regeneration of the cortex following long-term glucocorticoid therapy. We define the signaling and lineage relationships that underlie the regeneration process.
Collapse
Affiliation(s)
- Isabella Finco
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan 48109
| | - Antonio M. Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan 48109
| | - Gary D. Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan 48109
- Endocrine Oncology Program, Comprehensive Cancer Center, University of Michigan Health System, Ann Arbor, Michigan 48109
| |
Collapse
|
31
|
Death of adrenocortical cells during murine acute T. cruzi infection is not associated with TNF-R1 signaling but mostly with the type II pathway of Fas-mediated apoptosis. Brain Behav Immun 2017; 65:284-295. [PMID: 28666938 DOI: 10.1016/j.bbi.2017.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/22/2017] [Accepted: 05/26/2017] [Indexed: 01/28/2023] Open
Abstract
Earlier studies from our laboratory demonstrated that acute experimental Trypanosoma cruzi infection promotes an intense inflammation along with a sepsis-like dysregulated adrenal response characterized by normal levels of ACTH with raised glucocorticoid secretion. Inflammation was also known to result in adrenal cell apoptosis, which in turn may influence HPA axis uncoupling. To explore factors and pathways which may be involved in the apoptosis of adrenal cells, together with its impact on the functionality of the gland, we carried out a series of studies in mice lacking death receptors, such as TNF-R1 (C57BL/6-Tnfrsf1a tm1Imx or TNF-R1-/-) or Fas ligand (C57BL/6 Fas-deficient lpr mice), undergoing acute T. cruzi infection. Here we demonstrate that the late hypercorticosterolism seen in C57BL/6 mice during acute T. cruzi infection coexists with and hyperplasia and hypertrophy of zona fasciculata, paralleled by increased number of apoptotic cells. Apoptosis seems to be mediated mainly by the type II pathway of Fas-mediated apoptosis, which engages the mitochondrial pathway of apoptosis triggering the cytochrome c release to increase caspase-3 activation. Fas-induced apoptosis of adrenocortical cells is also related with an exacerbated production of intra-adrenal cytokines that probably maintain the late supply of adrenal hormones during host response. Present results shed light on the molecular mechanisms dealing with these phenomena which are crucial not only for the development of interventions attempting to avoid adrenal dysfunction, but also for its wide occurrence in other infectious-based critical illnesses.
Collapse
|
32
|
Menzies RI, Zhao X, Mullins LJ, Mullins JJ, Cairns C, Wrobel N, Dunbar DR, Bailey MA, Kenyon CJ. Transcription controls growth, cell kinetics and cholesterol supply to sustain ACTH responses. Endocr Connect 2017; 6:446-457. [PMID: 28720595 PMCID: PMC5574282 DOI: 10.1530/ec-17-0092] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 07/18/2017] [Indexed: 01/29/2023]
Abstract
Chronic ACTH exposure is associated with adrenal hypertrophy and steroidogenesis. The underlying molecular processes in mice have been analysed by microarray, histological and immunohistochemical techniques. Synacthen infused for 2 weeks markedly increased adrenal mass and plasma corticosterone levels. Microarray analysis found greater than 2-fold changes in expression of 928 genes (P < 0.001; 397 up, 531 down). These clustered in pathways involved in signalling, sterol/lipid metabolism, cell proliferation/hypertrophy and apoptosis. Signalling genes included some implicated in adrenal adenomas but also upregulated genes associated with cyclic AMP and downregulated genes associated with aldosterone synthesis. Sterol metabolism genes were those promoting cholesterol supply (Scarb1, Sqle, Apoa1) and disposal (Cyp27a1, Cyp7b1). Oil red O staining showed lipid depletion consistent with reduced expression of genes involved in lipid synthesis. Genes involved in steroidogenesis (Star, Cyp11a1, Cyp11b1) were modestly affected (P < 0.05; <1.3-fold). Increased Ki67, Ccna2, Ccnb2 and Tk1 expression complemented immunohistochemical evidence of a 3-fold change in cell proliferation. Growth arrest genes, Cdkn1a and Cdkn1c, which are known to be active in hypertrophied cells, were increased >4-fold and cross-sectional area of fasciculata cells was 2-fold greater. In contrast, genes associated with apoptosis (eg Casp12, Clu,) were downregulated and apoptotic cells (Tunel staining) were fewer (P < 0.001) and more widely distributed throughout the cortex. In summary, long-term steroidogenesis with ACTH excess is sustained by genes controlling cholesterol supply and adrenal mass. ACTH effects on adrenal morphology and genes controlling cell hypertrophy, proliferation and apoptosis suggest the involvement of different cell types and separate molecular pathways.
Collapse
Affiliation(s)
- Robert I Menzies
- The University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Xin Zhao
- The University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Linda J Mullins
- The University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - John J Mullins
- The University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Carolynn Cairns
- The University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Nicola Wrobel
- The University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Donald R Dunbar
- The University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Matthew A Bailey
- The University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Christopher J Kenyon
- The University/BHF Centre for Cardiovascular ScienceUniversity of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| |
Collapse
|
33
|
Comparison of two related lines of tauGFP transgenic mice designed for lineage tracing. BMC DEVELOPMENTAL BIOLOGY 2017; 17:8. [PMID: 28662681 PMCID: PMC5492368 DOI: 10.1186/s12861-017-0149-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/01/2017] [Indexed: 12/18/2022]
Abstract
Background The tauGFP reporter fusion protein is produced nearly ubiquitously by the TgTP6.3 transgene in TP6.3 mice and its localisation to microtubules offers some advantages over soluble GFP as a lineage marker. However, TgTP6.3Tg/Tg homozygotes are not viable and TgTP6.3Tg/− hemizygotes are smaller than wild-type. TP6.4 mice carry the TgTP6.4 transgene, which was produced with the same construct used to generate TgTP6.3, so we investigated whether TgTP6.4 had any advantages over TgTP6.3. Results Although TgTP6.4Tg/Tg homozygotes died before weaning, TgTP6.4Tg/− hemizygotes were viable and fertile and only males were significantly lighter than wild-type. The TgTP6.4 transgene produced the tauGFP fusion protein by the 2-cell stage and it was widely expressed in adults but tauGFP fluorescence was weak or absent in several tissues, including some neural tissues. The TgTP6.4 transgene expression pattern changed over several years of breeding and mosaic transgene expression became increasingly common in all expressing tissues. This mosaicism was used to visualise clonal lineages in the adrenal cortex of TgTP6.4Tg/− hemizygotes and these were qualitatively and quantitatively comparable to lineages reported previously for other mosaic transgenic mice, X-inactivation mosaics and chimaeras. Mosaicism occurred less frequently in TP6.3 than TP6.4 mice and was only observed in the corneal epithelium and adrenal cortex. Conclusions Mosaic expression makes the TgTP6.4 transgene unsuitable for use as a conventional cell lineage marker but such mosaicism provides a useful system for visualising clonal lineages that arise during development or maintenance of adult tissues. Differences in the occurrence of mosaicism between related transgenic lines, such as that described for lines TP6.3 and TP6.4, might provide a useful system for investigating the mechanism of transgene silencing. Electronic supplementary material The online version of this article (doi:10.1186/s12861-017-0149-x) contains supplementary material, which is available to authorized users.
Collapse
|
34
|
Steenblock C, Rubin de Celis MF, Androutsellis-Theotokis A, Sue M, Delgadillo Silva LF, Eisenhofer G, Andoniadou CL, Bornstein SR. Adrenal cortical and chromaffin stem cells: Is there a common progeny related to stress adaptation? Mol Cell Endocrinol 2017; 441:156-163. [PMID: 27637345 DOI: 10.1016/j.mce.2016.09.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/12/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022]
Abstract
The adrenal gland is a highly plastic organ with the capacity to adapt the body homeostasis to different physiological needs. The existence of stem-like cells in the adrenal cortex has been revealed in many studies. Recently, we identified and characterized in mice a pool of glia-like multipotent Nestin-expressing progenitor cells, which contributes to the plasticity of the adrenal medulla. In addition, we found that these Nestin progenitors are actively involved in the stress response by giving rise to chromaffin cells. Interestingly, we also observed a Nestin-GFP-positive cell population located under the adrenal capsule and scattered through the cortex. In this article, we discuss the possibility of a common progenitor giving rise to subpopulations of cells both in the adrenal cortex and medulla, the isolation and characterization of this progenitor as well as its clinical potential in transplantation therapies and in pathophysiology.
Collapse
Affiliation(s)
- Charlotte Steenblock
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany.
| | | | - Andreas Androutsellis-Theotokis
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany; Stem Cells, Tissue Engineering and Modelling (STEM), Division of Cancer and Stem Cells, University of Nottingham, Nottingham, UK
| | - Mariko Sue
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | | | - Graeme Eisenhofer
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Cynthia L Andoniadou
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany; Department of Craniofacial Development and Stem Cell Biology, King's College London, London, UK
| | - Stefan R Bornstein
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany; Department of Endocrinology and Diabetes, King's College London, London, UK
| |
Collapse
|
35
|
Pignatti E, Leng S, Carlone DL, Breault DT. Regulation of zonation and homeostasis in the adrenal cortex. Mol Cell Endocrinol 2017; 441:146-155. [PMID: 27619404 PMCID: PMC5235909 DOI: 10.1016/j.mce.2016.09.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/01/2016] [Accepted: 09/06/2016] [Indexed: 12/23/2022]
Abstract
The adult adrenal cortex is organized into concentric zones, each specialized to produce distinct steroid hormones. Cellular composition of the cortex is highly dynamic and subject to diverse signaling controls. Cortical homeostasis and regeneration rely on centripetal migration of steroidogenic cells from the outer to the inner cortex, which is accompanied by direct conversion of zona glomerulosa (zG) into zona fasciculata (zF) cells. Given the important impact of tissue structure and growth on steroidogenic function, it is essential to understand the mechanisms governing adrenal zonation and homeostasis. Towards this end, we review the distinctions between each zone by highlighting their morphological and ultra-structural features, discuss key signaling pathways influencing zonal identity, and evaluate current evidence for long-term self-renewing stem cells in the adult cortex. Finally, we review data supporting zG-to-zF transdifferentiation/direct conversion as a major mechanism of adult cortical renewal.
Collapse
Affiliation(s)
- Emanuele Pignatti
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Sining Leng
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA; Division of Medical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | - Diana L Carlone
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
36
|
Lerario AM, Finco I, LaPensee C, Hammer GD. Molecular Mechanisms of Stem/Progenitor Cell Maintenance in the Adrenal Cortex. Front Endocrinol (Lausanne) 2017; 8:52. [PMID: 28386245 PMCID: PMC5362593 DOI: 10.3389/fendo.2017.00052] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 03/03/2017] [Indexed: 01/10/2023] Open
Abstract
The adrenal cortex is characterized by three histologically and functionally distinct zones: the outermost zona glomerulosa (zG), the intermediate zona fasciculata, and the innermost zona reticularis. Important aspects of the physiology and maintenance of the adrenocortical stem/progenitor cells have emerged in the last few years. Studies have shown that the adrenocortical cells descend from a pool of progenitors that are localized in the subcapsular region of the zG. These cells continually undergo a process of centripetal displacement and differentiation, which is orchestrated by several paracrine and endocrine cues, including the pituitary-derived adrenocorticotrophic hormone, and angiotensin II. However, while several roles of the endocrine axes on adrenocortical function are well established, the mechanisms coordinating the maintenance of an undifferentiated progenitor cell pool with self-renewal capacity are poorly understood. Local factors, such as the composition of the extracellular matrix (ECM) with embedded signaling molecules, and the activity of major paracrine effectors, including ligands of the sonic hedgehog and Wnt signaling pathways, are thought to play a major role. Particularly, the composition of the ECM, which exhibits substantial differences within each of the three histologically distinct concentric zones, has been shown to influence the differentiation status of adrenocortical cells. New data from other organ systems and different experimental paradigms strongly support the conclusion that the interactions of ECM components with cell-surface receptors and secreted factors are key determinants of cell fate. In this review, we summarize established and emerging data on the paracrine and autocrine regulatory loops that regulate the biology of the progenitor cell niche and propose a role for bioengineered ECM models in further elucidating this biology in the adrenal.
Collapse
Affiliation(s)
- Antonio Marcondes Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Isabella Finco
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Christopher LaPensee
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Gary Douglas Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Endocrine Oncology Program, Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Center for Organogenesis, University of Michigan, Ann Arbor, MI, USA
- *Correspondence: Gary Douglas Hammer,
| |
Collapse
|
37
|
Vinson GP. Functional Zonation of the Adult Mammalian Adrenal Cortex. Front Neurosci 2016; 10:238. [PMID: 27378832 PMCID: PMC4908136 DOI: 10.3389/fnins.2016.00238] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/17/2016] [Indexed: 12/31/2022] Open
Abstract
The standard model of adrenocortical zonation holds that the three main zones, glomerulosa, fasciculata, and reticularis each have a distinct function, producing mineralocorticoids (in fact just aldosterone), glucocorticoids, and androgens respectively. Moreover, each zone has its specific mechanism of regulation, though ACTH has actions throughout. Finally, the cells of the cortex originate from a stem cell population in the outer cortex or capsule, and migrate centripetally, changing their phenotype as they progress through the zones. Recent progress in understanding the development of the gland and the distribution of steroidogenic enzymes, trophic hormone receptors, and other factors suggests that this model needs refinement. Firstly, proliferation can take place throughout the gland, and although the stem cells are certainly located in the periphery, zonal replenishment can take place within zones. Perhaps more importantly, neither the distribution of enzymes nor receptors suggest that the individual zones are necessarily autonomous in their production of steroid. This is particularly true of the glomerulosa, which does not seem to have the full suite of enzymes required for aldosterone biosynthesis. Nor, in the rat anyway, does it express MC2R to account for the response of aldosterone to ACTH. It is known that in development, recruitment of stem cells is stimulated by signals from within the glomerulosa. Furthermore, throughout the cortex local regulatory factors, including cytokines, catecholamines and the tissue renin-angiotensin system, modify and refine the effects of the systemic trophic factors. In these and other ways it more and more appears that the functions of the gland should be viewed as an integrated whole, greater than the sum of its component parts.
Collapse
Affiliation(s)
- Gavin P Vinson
- School of Biological and Chemical Sciences, Queen Mary University of London London, UK
| |
Collapse
|
38
|
Keighren MA, Flockhart JH, West JD. Survival of glucose phosphate isomerase null somatic cells and germ cells in adult mouse chimaeras. Biol Open 2016; 5:596-610. [PMID: 27103217 PMCID: PMC4874354 DOI: 10.1242/bio.017111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/22/2016] [Indexed: 01/22/2023] Open
Abstract
The mouse Gpi1 gene encodes the glycolytic enzyme glucose phosphate isomerase. Homozygous Gpi1(-/-) null mouse embryos die but a previous study showed that some homozygous Gpi1(-/-) null cells survived when combined with wild-type cells in fetal chimaeras. One adult female Gpi1(-/-)↔Gpi1(c/c) chimaera with functional Gpi1(-/-) null oocytes was also identified in a preliminary study. The aims were to characterise the survival of Gpi1(-/-) null cells in adult Gpi1(-/-)↔Gpi1(c/c) chimaeras and determine if Gpi1(-/-) null germ cells are functional. Analysis of adult Gpi1(-/-)↔Gpi1(c/c) chimaeras with pigment and a reiterated transgenic lineage marker showed that low numbers of homozygous Gpi1(-/-) null cells could survive in many tissues of adult chimaeras, including oocytes. Breeding experiments confirmed that Gpi1(-/-) null oocytes in one female Gpi1(-/-)↔Gpi1(c/c) chimaera were functional and provided preliminary evidence that one male putative Gpi1(-/-)↔Gpi1(c/c) chimaera produced functional spermatozoa from homozygous Gpi1(-/-) null germ cells. Although the male chimaera was almost certainly Gpi1(-/-)↔Gpi1(c/c), this part of the study is considered preliminary because only blood was typed for GPI. Gpi1(-/-) null germ cells should survive in a chimaeric testis if they are supported by wild-type Sertoli cells. It is also feasible that spermatozoa could bypass a block at GPI, but not blocks at some later steps in glycolysis, by using fructose, rather than glucose, as the substrate for glycolysis. Although chimaera analysis proved inefficient for studying the fate of Gpi1(-/-) null germ cells, it successfully identified functional Gpi1(-/-) null oocytes and revealed that some Gpi1(-/-) null cells could survive in many adult tissues.
Collapse
Affiliation(s)
- Margaret A Keighren
- Genes and Development Group, Centre for Integrative Physiology, Clinical Sciences, University of Edinburgh Medical School, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Jean H Flockhart
- Genes and Development Group, Centre for Integrative Physiology, Clinical Sciences, University of Edinburgh Medical School, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - John D West
- Genes and Development Group, Centre for Integrative Physiology, Clinical Sciences, University of Edinburgh Medical School, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| |
Collapse
|
39
|
Zilbermint M, Xekouki P, Faucz FR, Berthon A, Gkourogianni A, Schernthaner-Reiter MH, Batsis M, Sinaii N, Quezado MM, Merino M, Hodes A, Abraham SB, Libé R, Assié G, Espiard S, Drougat L, Ragazzon B, Davis A, Gebreab SY, Neff R, Kebebew E, Bertherat J, Lodish MB, Stratakis CA. Primary Aldosteronism and ARMC5 Variants. J Clin Endocrinol Metab 2015; 100:E900-9. [PMID: 25822102 PMCID: PMC4454793 DOI: 10.1210/jc.2014-4167] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
CONTEXT Primary aldosteronism is one of the leading causes of secondary hypertension, causing significant morbidity and mortality. A number of genetic defects have recently been identified in primary aldosteronism, whereas we identified mutations in ARMC5, a tumor-suppressor gene, in cortisol-producing primary macronodular adrenal hyperplasia. OBJECTIVE We investigated a cohort of 56 patients who were referred to the National Institutes of Health for evaluation of primary aldosteronism for ARMC5 defects. METHODS Patients underwent step-wise diagnosis, with measurement of serum aldosterone and plasma renin activity followed by imaging, saline suppression and/or oral salt loading tests, plus adrenal venous sampling. Cortisol secretion was also evaluated; unilateral or bilateral adrenalectomy was performed, if indicated. DNA, protein, and transfection studies in H295R cells were conducted by standard methods. RESULTS We identified 12 germline ARMC5 genetic alterations in 20 unrelated and two related individuals in our cohort (39.3%). ARMC5 sequence changes in 6 patients (10.7%) were predicted to be damaging by in silico analysis. All affected patients carrying a variant predicted to be damaging were African Americans (P = .0023). CONCLUSIONS Germline ARMC5 variants may be associated with primary aldosteronism. Additional cohorts of patients with primary aldosteronism and metabolic syndrome, particularly African Americans, should be screened for ARMC5 sequence variants because these may underlie part of the known increased predisposition of African Americans to low renin hypertension.
Collapse
|
40
|
Abstract
Aldosterone is a steroid hormone synthesized in and secreted from the outer layer of the adrenal cortex, the zona glomerulosa. Aldosterone is responsible for regulating sodium homeostasis, thereby helping to control blood volume and blood pressure. Insufficient aldosterone secretion can lead to hypotension and circulatory shock, particularly in infancy. On the other hand, excessive aldosterone levels, or those too high for sodium status, can cause hypertension and exacerbate the effects of high blood pressure on multiple organs, contributing to renal disease, stroke, visual loss, and congestive heart failure. Aldosterone is also thought to directly induce end-organ damage, including in the kidneys and heart. Because of the significance of aldosterone to the physiology and pathophysiology of the cardiovascular system, it is important to understand the regulation of its biosynthesis and secretion from the adrenal cortex. Herein, the mechanisms regulating aldosterone production in zona glomerulosa cells are discussed, with a particular emphasis on signaling pathways involved in the secretory response to the main controllers of aldosterone production, the renin-angiotensin II system, serum potassium levels and adrenocorticotrophic hormone. The signaling pathways involved include phospholipase C-mediated phosphoinositide hydrolysis, inositol 1,4,5-trisphosphate, cytosolic calcium levels, calcium influx pathways, calcium/calmodulin-dependent protein kinases, diacylglycerol, protein kinases C and D, 12-hydroxyeicostetraenoic acid, phospholipase D, mitogen-activated protein kinase pathways, tyrosine kinases, adenylate cyclase, and cAMP-dependent protein kinase. A complete understanding of the signaling events regulating aldosterone biosynthesis may allow the identification of novel targets for therapeutic interventions in hypertension, primary aldosteronism, congestive heart failure, renal disease, and other cardiovascular disorders.
Collapse
Affiliation(s)
- Wendy B Bollag
- Charlie Norwood VA Medical Center, Augusta, Georgia; Department of Physiology, Medical College of Georgia at Georgia Regents University, Augusta, Georgia
| |
Collapse
|
41
|
Abstract
Stem cells are endowed with the potential for self-renewal and multipotency. Pluripotent embryonic stem cells have an early role in the formation of the three germ layers (ectoderm, mesoderm and endoderm), whereas adult tissue stem cells and progenitor cells are critical mediators of organ homeostasis. The adrenal cortex is an exceptionally dynamic endocrine organ that is homeostatically maintained by paracrine and endocrine signals throughout postnatal life. In the past decade, much has been learned about the stem and progenitor cells of the adrenal cortex and the multiple roles that these cell populations have in normal development and homeostasis of the adrenal gland and in adrenal diseases. In this Review, we discuss the evidence for the presence of adrenocortical stem cells, as well as the various signalling molecules and transcriptional networks that are critical for the embryological establishment and postnatal maintenance of this vital population of cells. The implications of these pathways and cells in the pathophysiology of disease are also addressed.
Collapse
Affiliation(s)
- Elisabeth M Walczak
- Division of Nephrology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Gary D Hammer
- Center for Organogenesis, Alfred Taubman Biomedical Sciences Research Building, Room 1528, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| |
Collapse
|