A cell atlas of human adrenal cortex development and disease

The adrenal glands synthesize and release essential steroid hormones such as cortisol and aldosterone, but the mechanisms underlying human adrenal gland development are not fully understood. Here, we combined single-cell and bulk RNA-sequencing, spatial transcriptomics, immunohistochemistry and micro-focus computed tomography to investigate key aspects of adrenal development in the first 20 weeks of gestation. We demonstrate rapid adrenal growth and vascularization, with cell division in the outer definitive zone (DZ). Steroidogenic pathways favor androgen synthesis in the central fetal zone (FZ), but DZ capacity to synthesize cortisol and aldosterone develops with time. Core transcriptional regulators were identified, with a role for HOPX in the DZ. Potential ligand- receptor interactions between mesenchyme and adrenal cortex were seen (e.g., RSPO3/LGR4). Growth-promoting imprinted genes were enriched in the developing cortex (e.g. IGF2, PEG3). These findings reveal new aspects of human adrenal development, and have clinical implications for understanding primary adrenal insufficiency and related postnatal adrenal disorders, such as adrenal tumor development, steroid disorders and neonatal stress.

[1]  C. Flück,et al.  Development and function of the fetal adrenal , 2022, Reviews in Endocrine and Metabolic Disorders.

[2]  M. O’Reilly,et al.  The role for long-term use of dehydroepiandrosterone in adrenal insufficiency , 2022, Current opinion in endocrinology, diabetes, and obesity.

[3]  Mingyao Li,et al.  β-Hydroxybutyrate suppresses colorectal cancer , 2022, Nature.

[4]  H. Watari,et al.  The developmental origin and the specification of the adrenal cortex in humans and cynomolgus monkeys , 2022, bioRxiv.

[5]  M. Herbert,et al.  Single-cell roadmap of human gonadal development , 2021, Nature.

[6]  H. Frederiksen,et al.  The effects of selected inhibitors on human fetal adrenal steroidogenesis differs under basal and ACTH-stimulated conditions , 2021, BMC Medicine.

[7]  M. Geffner,et al.  Low Adrenomedullary Function Predicts Acute Illness in Infants with Classical Congenital Adrenal Hyperplasia. , 2021, The Journal of clinical endocrinology and metabolism.

[8]  A. Schedl,et al.  Adrenal cortex renewal in health and disease , 2021, Nature Reviews Endocrinology.

[9]  M. Dattani,et al.  Genetic Analysis of Pediatric Primary Adrenal Insufficiency of Unknown Etiology: 25 Years’ Experience in the UK , 2021, Journal of the Endocrine Society.

[10]  O. Arthurs,et al.  Human fetal whole-body postmortem microfocus computed tomographic imaging , 2021, Nature Protocols.

[11]  A. Belgorosky,et al.  Normal and Premature Adrenarche. , 2021, Endocrine reviews.

[12]  S. Bornstein,et al.  Adrenal medulla development and medullary-cortical interactions , 2021, Molecular and Cellular Endocrinology.

[13]  M. Monteiro,et al.  Angiogenesis in the Normal Adrenal Fetal Cortex and Adrenocortical Tumors , 2021, Cancers.

[14]  C. Flück,et al.  Adrenal cortex development and related disorders leading to adrenal insufficiency , 2021, Molecular and Cellular Endocrinology.

[15]  G. Hammer,et al.  β-catenin in adrenal zonation and disease , 2020, Molecular and Cellular Endocrinology.

[16]  Marina Sirota,et al.  dittoSeq: universal user-friendly single-cell and bulk RNA sequencing visualization toolkit , 2020, Bioinform..

[17]  G. Hammer,et al.  Stem cell function and plasticity in the normal physiology of the adrenal cortex , 2020, Molecular and Cellular Endocrinology.

[18]  V. Rangnekar,et al.  Lessons from Mouse Models , 2021, Tumor Suppressor Par-4.

[19]  Lee B. Smith,et al.  Androgen Receptor Is Dispensable for X-Zone Regression in the Female Adrenal but Regulates Post-Partum Corticosterone Levels and Protects Cortex Integrity , 2021, Frontiers in Endocrinology.

[20]  I. del Valle,et al.  Current Insights Into Adrenal Insufficiency in the Newborn and Young Infant , 2020, Frontiers in Pediatrics.

[21]  H. Frederiksen,et al.  Establishment of a novel human fetal adrenal culture model that supports de novo and manipulated steroidogenesis. , 2020, The Journal of clinical endocrinology and metabolism.

[22]  V. Morelli,et al.  The cytoskeleton actin binding protein filamin A impairs both IGF2 mitogenic effects and the efficacy of IGF1R inhibitors in adrenocortical cancer cells. , 2020, Cancer letters.

[23]  Yidong Chen,et al.  Knock-out of Hopx disrupts stemness and quiescence of hematopoietic stem cells in mice , 2020, Oncogene.

[24]  David A. Williams,et al.  The Changing Face of Adrenoleukodystrophy. , 2020, Endocrine reviews.

[25]  R. Auchus,et al.  11-Oxygenated androgens in health and disease , 2020, Nature Reviews Endocrinology.

[26]  L. Sibley,et al.  Long-Term Culture Captures Injury-Repair Cycles of Colonic Stem Cells , 2019, Cell.

[27]  Lee B. Smith,et al.  Androgen receptor signalling in the male adrenal facilitates X-zone regression, cell turnover and protects against adrenal degeneration during ageing , 2019, Scientific Reports.

[28]  K. Storbeck,et al.  Steroid Metabolome Analysis in Disorders of Adrenal Steroid Biosynthesis and Metabolism , 2019, Endocrine reviews.

[29]  W. Drake,et al.  DLK1/PREF1 marks a novel cell population in the human adrenal cortex , 2019, The Journal of Steroid Biochemistry and Molecular Biology.

[30]  Francisca Rojas Ringeling,et al.  A Common Embryonic Origin of Stem Cells Drives Developmental and Adult Neurogenesis , 2019, Cell.

[31]  N. Hanley,et al.  Alternative pathway androgen biosynthesis and human fetal female virilization , 2019, Proceedings of the National Academy of Sciences.

[32]  R. Nusse,et al.  A ZNRF3-dependent Wnt/β-catenin signaling gradient is required for adrenal homeostasis , 2019, Genes & development.

[33]  T. Eggermann,et al.  Genomic imprinting disorders: lessons on how genome, epigenome and environment interact , 2019, Nature Reviews Genetics.

[34]  H. Frederiksen,et al.  Characterization of Human Adrenal Steroidogenesis During Fetal Development , 2018, The Journal of clinical endocrinology and metabolism.

[35]  M. Weger,et al.  Expression and activity profiling of the steroidogenic enzymes of glucocorticoid biosynthesis and the fdx1 co‐factors in zebrafish , 2018, Journal of neuroendocrinology.

[36]  Luke Zappia,et al.  Clustering trees: a visualization for evaluating clusterings at multiple resolutions , 2018, bioRxiv.

[37]  M. Simard,et al.  The human fetal adrenal produces cortisol but no detectable aldosterone throughout the second trimester , 2018, BMC Medicine.

[38]  V. Ducros,et al.  PKA signaling drives reticularis differentiation and sexually dimorphic adrenal cortex renewal. , 2018, JCI insight.

[39]  Adam A. Margolin,et al.  Chromatin and Transcriptional Analysis of Mesoderm Progenitor Cells Identifies HOPX as a Regulator of Primitive Hematopoiesis. , 2017, Cell reports.

[40]  Lin Lin,et al.  A genomic atlas of human adrenal and gonad development , 2017, Wellcome open research.

[41]  J. Schwabe,et al.  Genetic disorders of nuclear receptors , 2017, The Journal of clinical investigation.

[42]  G. Kokai,et al.  Somatic mutations and progressive monosomy modify SAMD9-related phenotypes in humans , 2017, The Journal of clinical investigation.

[43]  G. Hammer,et al.  Molecular Mechanisms of Stem/Progenitor Cell Maintenance in the Adrenal Cortex , 2017, Front. Endocrinol..

[44]  Xinran Tang,et al.  HOPX hypermethylation promotes metastasis via activating SNAIL transcription in nasopharyngeal carcinoma , 2017, Nature Communications.

[45]  K. Yamashita,et al.  Epigenetic silencing of HOPX contributes to cancer aggressiveness in breast cancer. , 2017, Cancer letters.

[46]  K. Yamashita,et al.  Homeobox-Only Protein Expression Is a Critical Prognostic Indicator of Pancreatic Neuroendocrine Tumor and Is Regulated by Promoter DNA Hypermethylation. , 2016, Pancreas.

[47]  V. Vidal,et al.  The adrenal capsule is a signaling center controlling cell renewal and zonation through Rspo3 , 2016, Genes & development.

[48]  David Taieb,et al.  Copy number variations alter methylation and parallel IGF2 overexpression in adrenal tumors , 2015, Endocrine-related cancer.

[49]  A. Duncan,et al.  DAX-1 (NR0B1) and steroidogenic factor-1 (SF-1, NR5A1) in human disease , 2015, Best practice & research. Clinical endocrinology & metabolism.

[50]  G. Hammer,et al.  Development of adrenal cortex zonation. , 2015, Endocrinology and metabolism clinics of North America.

[51]  J. Epstein,et al.  Plasticity of Hopx+ Type I alveolar cells to regenerate Type II cells in the lung , 2015, Nature Communications.

[52]  G. Hammer,et al.  Regulation of the adrenocortical stem cell niche: implications for disease , 2015, Nature Reviews Endocrinology.

[53]  H. Sasano,et al.  Transcriptome profiling reveals differentially expressed transcripts between the human adrenal zona fasciculata and zona reticularis. , 2014, The Journal of clinical endocrinology and metabolism.

[54]  G. Hammer,et al.  Fetal adrenal capsular cells serve as progenitor cells for steroidogenic and stromal adrenocortical cell lineages in M. musculus , 2013, Development.

[55]  P. Barrett,et al.  Adrenocortical zonation results from lineage conversion of differentiated zona glomerulosa cells. , 2013, Developmental cell.

[56]  S. Nelson,et al.  Mutations in the PCNA-binding domain of CDKN1C cause IMAGE Syndrome , 2012, Nature Genetics.

[57]  L. Metherell,et al.  MCM4 mutation causes adrenal failure, short stature, and natural killer cell deficiency in humans. , 2012, The Journal of clinical investigation.

[58]  R. Jaffe,et al.  Development and function of the human fetal adrenal cortex: a key component in the feto-placental unit. , 2011, Endocrine reviews.

[59]  E. Lalli,et al.  Genetics and genomics of childhood adrenocortical tumors , 2011, Molecular and Cellular Endocrinology.

[60]  Sonia Shah,et al.  ChIP-on-chip analysis reveals angiopoietin 2 (Ang2, ANGPT2) as a novel target of steroidogenic factor-1 (SF-1, NR5A1) in the human adrenal gland , 2011, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[61]  W. Miller,et al.  The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. , 2011, Endocrine reviews.

[62]  W. Rainey,et al.  ACTH is a potent regulator of gene expression in human adrenal cells. , 2010, Journal of molecular endocrinology.

[63]  R. Kuick,et al.  Preclinical targeting of the type I insulin-like growth factor receptor in adrenocortical carcinoma. , 2009, The Journal of clinical endocrinology and metabolism.

[64]  R. Jaffe,et al.  The periphery of the human fetal adrenal gland is a site of angiogenesis: zonal differential expression and regulation of angiogenic factors. , 2008, The Journal of clinical endocrinology and metabolism.

[65]  E. Lalli,et al.  Nephroblastoma overexpressed/cysteine-rich protein 61/connective tissue growth factor/nephroblastoma overexpressed gene-3 (NOV/CCN3), a selective adrenocortical cell proapoptotic factor, is down-regulated in childhood adrenocortical tumors. , 2007, The Journal of clinical endocrinology and metabolism.

[66]  E. Lalli,et al.  SF-1 overexpression in childhood adrenocortical tumours. , 2006, European journal of cancer.

[67]  David I. Wilson,et al.  In humans, early cortisol biosynthesis provides a mechanism to safeguard female sexual development. , 2006, The Journal of clinical investigation.

[68]  A. Belgorosky,et al.  Expression of the IGF System in Human Adrenal Tissues from Early Infancy to Late Puberty: Implications for the Development of Adrenarche , 2005, Pediatric Research.

[69]  E. Lalli,et al.  Amplification of the steroidogenic factor 1 gene in childhood adrenocortical tumors. , 2005, The Journal of clinical endocrinology and metabolism.

[70]  S. Bornstein,et al.  Cortical-chromaffin cell interactions in the adrenal gland , 2005, Endocrine pathology.

[71]  R. Jaffe,et al.  Identification of definitive and fetal zone markers in the human fetal adrenal gland reveals putative developmental genes. , 2003, The Journal of clinical endocrinology and metabolism.

[72]  G. Chrousos,et al.  Adrenomedullary function may predict phenotype and genotype in classic 21-hydroxylase deficiency. , 2002, The Journal of clinical endocrinology and metabolism.

[73]  W. Rainey,et al.  Gene profiling of human fetal and adult adrenals. , 2001, The Journal of endocrinology.

[74]  S. Hennig,et al.  Triple A syndrome is caused by mutations in AAAS, a new WD-repeat protein gene. , 2001, Human molecular genetics.

[75]  C. Deal,et al.  Pediatric adrenocortical tumors: molecular events leading to insulin-like growth factor II gene overexpression. , 2000, The Journal of clinical endocrinology and metabolism.

[76]  David I. Wilson,et al.  Expression of steroidogenic factor 1 and Wilms' tumour 1 during early human gonadal development and sex determination , 1999, Mechanisms of Development.

[77]  G. Vinson,et al.  Pref-1, SF-1 and adrenocortical zonation. , 1998, Endocrine research.

[78]  M. Okamoto,et al.  Cloning of a membrane-spanning protein with epidermal growth factor-like repeat motifs from adrenal glomerulosa cells. , 1998, Endocrinology.

[79]  S. Mesiano,et al.  Developmental and functional biology of the primate fetal adrenal cortex. , 1997, Endocrine reviews.

[80]  M. Ito,et al.  DAX-1 inhibits SF-1-mediated transactivation via a carboxy-terminal domain that is deleted in adrenal hypoplasia congenita , 1997, Molecular and cellular biology.

[81]  R. Carey,et al.  Adrenal insufficiency. , 1997, Current therapy in endocrinology and metabolism.

[82]  A. Monaco,et al.  Mutations in the DAX-1 gene give rise to both X-linked adrenal hypoplasia congenita and hypogonadotropic hypogonadism , 1994, Nature.

[83]  K. Parker,et al.  A cell-specific nuclear receptor is essential for adrenal and gonadal development and sexual differentiation , 1994, Cell.

[84]  A. Hochberg,et al.  Parallel regulation of parentally imprinted H19 and insulin-like growth factor-II genes in cultured human fetal adrenal cells. , 1994, Endocrinology.