137 results on '"Hammer GD"'
Search Results
2. Tissue-Microarray of Beta-Catenin in a Cohort of Pediatric and Adult Adrenocortical Tumors.
- Author
-
Lima, LO, primary, Soares, IC, additional, Almeida, MQ, additional, Fragoso, MCBV, additional, Lerario, AM, additional, Ribeiro, TC, additional, Mariani, TC, additional, Hammer, GD, additional, Mendonca, BB, additional, and Latronico, AC, additional
- Published
- 2010
- Full Text
- View/download PDF
3. p53 deficiency partially rescues the phenotype of the acd mouse at the expense of increased carcinogenesis
- Author
-
Else, T, primary and Hammer, GD, additional
- Published
- 2007
- Full Text
- View/download PDF
4. Unravelling the molecular phenotype of adrenocortical dysplasia (acd) mice
- Author
-
Else, T, primary, Theisen, BK, additional, Keegan, CE, additional, Ferguson, DO, additional, and Hammer, GD, additional
- Published
- 2006
- Full Text
- View/download PDF
5. Agouti Related Peptide (AgRP) deficient mice show accelerated compensatory adrenal growth after unilateral adrenalectomy
- Author
-
Else, T, primary, Kim, AC, additional, and Hammer, GD, additional
- Published
- 2005
- Full Text
- View/download PDF
6. The Role of Inhibin and Activin in Adrenal Tumorigenesis
- Author
-
Beuschlein, F, primary, Looyenga, BD, additional, and Hammer, GD, additional
- Published
- 2004
- Full Text
- View/download PDF
7. Gonadectomized mice of the inbred strain CE/J develop adrenocortical tumors derived from subcapsular cells expressing x-zone marker
- Author
-
Beuschlein, F, primary, Shapiro, I, additional, Hammer, GD, additional, and Reincke, M, additional
- Published
- 2004
- Full Text
- View/download PDF
8. Non-canonical Wnt signaling triggered by WNT2B drives adrenal aldosterone production.
- Author
-
Borges KS, Little DW 3rd, Magalhães TA, Ribeiro C, Dumontet T, Lapensee C, Basham KJ, Seth A, Azova S, Guagliardo NA, Barrett PQ, Berber M, O'Connell AE, Turcu AF, Lerario AM, Mohan DR, Rainey W, Carlone DL, Hirschhorn JN, Salic A, Breault DT, and Hammer GD
- Abstract
The steroid hormone aldosterone, produced by the zona glomerulosa (zG) of the adrenal gland, is a master regulator of plasma electrolytes and blood pressure. While aldosterone control by the renin-angiotensin system is well understood, other key regulatory factors have remained elusive. Here, we replicated a prior association between a non-coding variant in WNT2B and an increased risk of primary aldosteronism, a prevalent and debilitating disease caused by excessive aldosterone production. We further show that in both mice and humans, WNT2B is expressed in the mesenchymal capsule surrounding the adrenal cortex, in close proximity to the zG. Global loss of Wnt2b in the mouse results in a dysmorphic and hypocellular zG, with impaired aldosterone production. Similarly, humans harboring WNT2B loss-of-function mutations develop a novel form of Familial Hyperreninemic Hypoaldosteronism, designated here as Type 4. Additionally, we demonstrate that WNT2B signals by activating the non-canonical Wnt/planar cell polarity pathway. Our findings identify WNT2B as a key regulator of zG function and aldosterone production with important clinical implications.
- Published
- 2024
- Full Text
- View/download PDF
9. EndoBridge 2023: highlights and pearls.
- Author
-
Yildiz BO, Boguszewski CL, da Silva Boguszewski MC, Busetto L, Celik O, Fuleihan GE, Goulis DG, Hammer GD, Haymart MR, Kaltsas G, Law JR, Lim AYL, Luger A, Macut D, McGowan B, McClung M, Miras AD, Patti ME, Peeters RP, Pignatelli D, Saeed H, Sipos J, Stratakis CA, Tsoli M, van der Lely AJ, Witchel SF, and Yazici D
- Subjects
- Humans, Endocrinology history, Osteoporosis therapy, Endocrine System Diseases therapy
- Abstract
EndoBridge 2023 took place on October 20-22, 2023, in Antalya, Turkey. Accredited by the European Council, the 3-day scientific program of the 11
th Annual Meeting of EndoBridge included state-of-the-art lectures and interactive small group discussion sessions incorporating interesting and challenging clinical cases led by globally recognized leaders in the field and was well attended by a highly diverse audience. Following its established format over the years, the program provided a comprehensive update across all aspects of endocrinology and metabolism, including topics in pituitary, thyroid, bone, and adrenal disorders, neuroendocrine tumors, diabetes mellitus, obesity, nutrition, and lipid disorders. As usual, the meeting was held in English with simultaneous translation into Russian, Arabic, and Turkish. The abstracts of clinical cases presented by the delegates during oral and poster sessions have been published in JCEM Case Reports. Herein, we provide a paper on highlights and pearls of the meeting sessions covering a wide range of subjects, from thyroid nodule stratification to secondary osteoporosis and from glycemic challenges in post-bariatric surgery to male hypogonadism. This report emphasizes the latest developments in the field, along with clinical approaches to common endocrine issues. The 12th annual meeting of EndoBridge will be held on October 17-20, 2024 in Antalya, Turkey., (© 2024. The Author(s), under exclusive licence to Hellenic Endocrine Society.)- Published
- 2024
- Full Text
- View/download PDF
10. Chronic activation of adrenal Gq signaling induces Cyp11b2 expression in the zona fasciculata and hyperaldosteronism.
- Author
-
van Rooyen D, Lerario AM, Little DW 3rd, Ullenbruch MR, Taylor MJ, Gomez-Sanchez CE, Hammer GD, and Rainey WE
- Subjects
- Male, Female, Humans, Mice, Animals, Zona Fasciculata, Aldosterone metabolism, Zona Glomerulosa metabolism, Cytochrome P-450 CYP11B2 genetics, Wnt Signaling Pathway, Mice, Transgenic, Adrenal Cortex metabolism, Hyperaldosteronism
- Abstract
Hyperaldosteronism is often associated with inappropriate aldosterone production and aldosterone synthase (Cyp11b2) expression. Normally, Cyp11b2 expression is limited to the adrenal zona glomerulosa (ZG) and regulated by angiotensin II which signals through Gq protein-coupled receptors. As cells migrate inwards, they differentiate into 11β-hydroxylase-expressing zona fasciculata (ZF) cells lacking Cyp11b2. The mechanism causing ZG-specific aldosterone biosynthesis is still unclear. We investigated the effect of chronic Gq signaling using transgenic mice with a clozapine N-oxide (CNO)-activated human M3 muscarinic receptor (DREADD) coupled to Gq (hM3Dq) that was expressed throughout the adrenal cortex. CNO raised circulating aldosterone in the presence of a high sodium diet with greater response seen in females compared to males. Immunohistochemistry and transcriptomics indicated disrupted zonal Cyp11b2 expression while Wnt signaling remained unchanged. Chronic Gq-DREADD signaling also induced an intra-adrenal RAAS in CNO-treated mice. Chronic Gq signaling disrupted adrenal cortex zonal aldosterone production associated with ZF expression of Cyp11b2., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. Potential involvement of the bone marrow in experimental Graves' disease and thyroid eye disease.
- Author
-
Gulbins A, Horstmann M, Keitsch S, Soddemann M, Wilker B, Wilson GC, Zeidan R, Hammer GD, Daser A, Bechrakis NE, Görtz GE, and Eckstein A
- Subjects
- Mice, Animals, Arginase, Bone Marrow metabolism, Receptors, Thyrotropin, Arginine, Graves Ophthalmopathy metabolism, Graves Disease, Autoimmune Diseases complications
- Abstract
Introduction: Graves' disease is an autoimmune disorder caused by auto-antibodies against the thyroid stimulating hormone receptor (TSHR). Overstimulation of the TSHR induces hyperthyroidism and thyroid eye disease (TED) as the most common extra thyroidal manifestation of Graves' disease. In TED, the TSHR cross talks with the insulin-like growth factor 1 receptor (IGF-1R) in orbital fibroblasts leading to inflammation, deposition of hyaluronan and adipogenesis. The bone marrow may play an important role in autoimmune diseases, but its role in Graves' disease and TED is unknown. Here, we investigated whether induction of experimental Graves' disease and accompanying TED involves bone marrow activation and whether interference with IGF-1R signaling prevents this activation., Results: Immunization of mice with TSHR resulted in an increase the numbers of CD4-positive T-lymphocytes (p ≤0.0001), which was normalized by linsitinib (p = 0.0029), an increase of CD19-positive B-lymphocytes (p= 0.0018), which was unaffected by linsitinib and a decrease of GR1-positive cells (p= 0.0038), which was prevented by linsitinib (p= 0.0027). In addition, we observed an increase of Sca-1 positive hematopietic stem cells (p= 0.0007) and of stromal cell-derived factor 1 (SDF-1) (p ≤0.0001) after immunization with TSHR which was prevented by linsitinib (Sca-1: p= 0.0008, SDF-1: p ≤0.0001). TSHR-immunization also resulted in upregulation of CCL-5, IL-6 and osteopontin (all p ≤0.0001) and a concomitant decrease of the immune-inhibitory cytokines IL-10 (p= 0.0064) and PGE2 (p ≤0.0001) in the bone marrow (all p≤ 0.0001). Treatment with the IGF-1R antagonist linsitinib blocked these events (all p ≤0.0001). We further demonstrate a down-regulation of arginase-1 expression (p= 0.0005) in the bone marrow in TSHR immunized mice, with a concomitant increase of local arginine (p ≤0.0001). Linsitinib induces an upregulation of arginase-1 resulting in low arginase levels in the bone marrow. Reconstitution of arginine in bone marrow cells in vitro prevented immune-inhibition by linsitinib., Conclusion: Collectively, these data indicate that the bone marrow is activated in experimental Graves' disease and TED, which is prevented by linsitinib. Linsitinib-mediated immune-inhibition is mediated, at least in part, by arginase-1 up-regulation, consumption of arginine and thereby immune inhibition., Competing Interests: Author RZ was employed by the company Sling Therapeutics Inc. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Gulbins, Horstmann, Keitsch, Soddemann, Wilker, Wilson, Zeidan, Hammer, Daser, Bechrakis, Görtz and Eckstein.)
- Published
- 2023
- Full Text
- View/download PDF
12. Targeting Oncogenic Wnt/β-Catenin Signaling in Adrenocortical Carcinoma Disrupts ECM Expression and Impairs Tumor Growth.
- Author
-
Penny MK, Lerario AM, Basham KJ, Chukkapalli S, Mohan DR, LaPensee C, Converso-Baran K, Hoenerhoff MJ, Suárez-Fernández L, Rey CGD, Giordano TJ, Han R, Newman EA, and Hammer GD
- Abstract
Adrenocortical carcinoma (ACC) is a rare but highly aggressive cancer with limited treatment options and poor survival for patients with advanced disease. An improved understanding of the transcriptional programs engaged in ACC will help direct rational, targeted therapies. Whereas activating mutations in Wnt/β-catenin signaling are frequently observed, the β-catenin-dependent transcriptional targets that promote tumor progression are poorly understood. To address this question, we analyzed ACC transcriptome data and identified a novel Wnt/β-catenin-associated signature in ACC enriched for the extracellular matrix (ECM) and predictive of poor survival. This suggested an oncogenic role for Wnt/β-catenin in regulating the ACC microenvironment. We further investigated the minor fibrillar collagen, collagen XI alpha 1 (COL11A1), and found that COL11A1 expression originates specifically from cancer cells and is strongly correlated with both Wnt/β-catenin activation and poor patient survival. Inhibition of constitutively active Wnt/β-catenin signaling in the human ACC cell line, NCI-H295R, significantly reduced the expression of COL11A1 and other ECM components and decreased cancer cell viability. To investigate the preclinical potential of Wnt/β-catenin inhibition in the adrenal microenvironment, we developed a minimally invasive orthotopic xenograft model of ACC and demonstrated that treatment with the newly developed Wnt/β-catenin:TBL1 inhibitor Tegavivint significantly reduced tumor growth. Together, our data support that the inhibition of aberrantly active Wnt/β-catenin disrupts transcriptional reprogramming of the microenvironment and reduces ACC growth and survival. Furthermore, this β-catenin-dependent oncogenic program can be therapeutically targeted with a newly developed Wnt/β-catenin inhibitor. These results show promise for the further clinical development of Wnt/β-catenin inhibitors in ACC and unveil a novel Wnt/β-catenin-regulated transcriptome.
- Published
- 2023
- Full Text
- View/download PDF
13. β-Catenin-Driven Differentiation Is a Tissue-Specific Epigenetic Vulnerability in Adrenal Cancer.
- Author
-
Mohan DR, Borges KS, Finco I, LaPensee CR, Rege J, Solon AL, Little DW, Else T, Almeida MQ, Dang D, Haggerty-Skeans J, Apfelbaum AA, Vinco M, Wakamatsu A, Mariani BMP, Amorim LC, Latronico AC, Mendonca BB, Zerbini MCN, Lawlor ER, Ohi R, Auchus RJ, Rainey WE, Marie SKN, Giordano TJ, Venneti S, Fragoso MCBV, Breault DT, Lerario AM, and Hammer GD
- Subjects
- Humans, beta Catenin genetics, beta Catenin metabolism, Epigenesis, Genetic, Chromatin genetics, Adrenocortical Carcinoma genetics, Adrenocortical Carcinoma metabolism, Adrenocortical Carcinoma pathology, Adrenal Cortex Neoplasms genetics, Adrenal Cortex Neoplasms pathology
- Abstract
Adrenocortical carcinoma (ACC) is a rare cancer in which tissue-specific differentiation is paradoxically associated with dismal outcomes. The differentiated ACC subtype CIMP-high is prevalent, incurable, and routinely fatal. CIMP-high ACC possess abnormal DNA methylation and frequent β-catenin-activating mutations. Here, we demonstrated that ACC differentiation is maintained by a balance between nuclear, tissue-specific β-catenin-containing complexes, and the epigenome. On chromatin, β-catenin bound master adrenal transcription factor SF1 and hijacked the adrenocortical super-enhancer landscape to maintain differentiation in CIMP-high ACC; off chromatin, β-catenin bound histone methyltransferase EZH2. SF1/β-catenin and EZH2/β-catenin complexes present in normal adrenals persisted through all phases of ACC evolution. Pharmacologic EZH2 inhibition in CIMP-high ACC expelled SF1/β-catenin from chromatin and favored EZH2/β-catenin assembly, erasing differentiation and restraining cancer growth in vitro and in vivo. These studies illustrate how tissue-specific programs shape oncogene selection, surreptitiously encoding targetable therapeutic vulnerabilities., Significance: Oncogenic β-catenin can use tissue-specific partners to regulate cellular differentiation programs that can be reversed by epigenetic therapies, identifying epigenetic control of differentiation as a viable target for β-catenin-driven cancers., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
14. Senescence-induced immune remodeling facilitates metastatic adrenal cancer in a sex-dimorphic manner.
- Author
-
Warde KM, Smith LJ, Liu L, Stubben CJ, Lohman BK, Willett PW, Ammer JL, Castaneda-Hernandez G, Imodoye SO, Zhang C, Jones KD, Converso-Baran K, Ekiz HA, Barry M, Clay MR, Kiseljak-Vassiliades K, Giordano TJ, Hammer GD, and Basham KJ
- Subjects
- Male, Animals, Female, Aging, Cellular Senescence, Signal Transduction, Tumor Microenvironment, Adrenocortical Carcinoma genetics, Adrenal Cortex Neoplasms genetics
- Abstract
Aging markedly increases cancer risk, yet our mechanistic understanding of how aging influences cancer initiation is limited. Here we demonstrate that the loss of ZNRF3, an inhibitor of Wnt signaling that is frequently mutated in adrenocortical carcinoma, leads to the induction of cellular senescence that remodels the tissue microenvironment and ultimately permits metastatic adrenal cancer in old animals. The effects are sexually dimorphic, with males exhibiting earlier senescence activation and a greater innate immune response, driven in part by androgens, resulting in high myeloid cell accumulation and lower incidence of malignancy. Conversely, females present a dampened immune response and increased susceptibility to metastatic cancer. Senescence-recruited myeloid cells become depleted as tumors progress, which is recapitulated in patients in whom a low myeloid signature is associated with worse outcomes. Our study uncovers a role for myeloid cells in restraining adrenal cancer with substantial prognostic value and provides a model for interrogating pleiotropic effects of cellular senescence in cancer., (© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
15. Linsitinib, an IGF-1R inhibitor, attenuates disease development and progression in a model of thyroid eye disease.
- Author
-
Gulbins A, Horstmann M, Daser A, Flögel U, Oeverhaus M, Bechrakis NE, Banga JP, Keitsch S, Wilker B, Krause G, Hammer GD, Spencer AG, Zeidan R, Eckstein A, Philipp S, and Görtz GE
- Subjects
- Animals, Mice, Hyperthyroidism, Imidazoles, Graves Disease drug therapy, Graves Ophthalmopathy drug therapy, Protein Kinase Inhibitors therapeutic use, Receptor, IGF Type 1 antagonists & inhibitors
- Abstract
Introduction: Graves' disease (GD) is an autoimmune disorder caused by autoantibodies against the thyroid stimulating hormone receptor (TSHR) leading to overstimulation of the thyroid gland. Thyroid eye disease (TED) is the most common extra thyroidal manifestation of GD. Therapeutic options to treat TED are very limited and novel treatments need to be developed. In the present study we investigated the effect of linsitinib, a dual small-molecule kinase inhibitor of the insulin-like growth factor 1 receptor (IGF-1R) and the Insulin receptor (IR) on the disease outcome of GD and TED., Methods: Linsitinib was administered orally for four weeks with therapy initiating in either the early ("active") or the late ("chronic") phases of the disease. In the thyroid and the orbit, autoimmune hyperthyroidism and orbitopathy were analyzed serologically (total anti-TSHR binding antibodies, stimulating anti TSHR antibodies, total T4 levels), immunohistochemically (H&E-, CD3-, TNFa- and Sirius red staining) and with immunofluorescence (F4/80 staining). An MRI was performed to quantify in vivo tissue remodeling inside the orbit., Results: Linsitinib prevented autoimmune hyperthyroidism in the early state of the disease, by reducing morphological changes indicative for hyperthyroidism and blocking T-cell infiltration, visualized by CD3 staining. In the late state of the disease linsitinib had its main effect in the orbit. Linsitinib reduced immune infiltration of T-cells (CD3 staining) and macrophages (F4/80 and TNFa staining) in the orbita in experimental GD suggesting an additional, direct effect of linsitinib on the autoimmune response. In addition, treatment with linsitinib normalized the amount of brown adipose tissue in both the early and late group. An in vivo MRI of the late group was performed and revealed a marked decrease of inflammation, visualized by
19 F MR imaging, significant reduction of existing muscle edema and formation of brown adipose tissue., Conclusion: Here, we demonstrate that linsitinib effectively prevents development and progression of thyroid eye disease in an experimental murine model for Graves' disease. Linsitinib improved the total disease outcome, indicating the clinical significance of the findings and providing a path to therapeutic intervention of Graves' Disease. Our data support the use of linsitinib as a novel treatment for thyroid eye disease., Competing Interests: Authors AS and RZ were employed by the company Sling Therapeutics Inc. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Gulbins, Horstmann, Daser, Flögel, Oeverhaus, Bechrakis, Banga, Keitsch, Wilker, Krause, Hammer, Spencer, Zeidan, Eckstein, Philipp and Görtz.)- Published
- 2023
- Full Text
- View/download PDF
16. Novel Candidate Regulators and Developmental Trajectory of Pituitary Thyrotropes.
- Author
-
Cheung LYM, Menage L, Rizzoti K, Hamilton G, Dumontet T, Basham K, Daly AZ, Brinkmeier ML, Masser BE, Treier M, Cobb J, Delogu A, Lovell-Badge R, Hammer GD, and Camper SA
- Subjects
- Pregnancy, Female, Mice, Animals, Thyrotropin metabolism, Pituitary Gland metabolism, Transcription Factors metabolism, Immunohistochemistry, SOXB2 Transcription Factors metabolism, Pituitary Diseases metabolism, Pituitary Gland, Anterior metabolism
- Abstract
The pituitary gland regulates growth, metabolism, reproduction, the stress response, uterine contractions, lactation, and water retention. It secretes hormones in response to hypothalamic input, end organ feedback, and diurnal cues. The mechanisms by which pituitary stem cells are recruited to proliferate, maintain quiescence, or differentiate into specific cell types, especially thyrotropes, are not well understood. We used single-cell RNA sequencing in juvenile P7 mouse pituitary cells to identify novel factors in pituitary cell populations, with a focus on thyrotropes and rare subtypes. We first observed cells coexpressing markers of both thyrotropes and gonadotropes, such as Pou1f1 and Nr5a1. This was validated in vivo by both immunohistochemistry and lineage tracing of thyrotropes derived from Nr5a1-Cre; mTmG mice and demonstrates that Nr5a1-progenitors give rise to a proportion of thyrotropes during development. Our data set also identifies novel factors expressed in pars distalis and pars tuberalis thyrotropes, including the Shox2b isoform in all thyrotropes and Sox14 specifically in Pou1f1-negative pars tuberalis thyrotropes. We have therefore used single-cell transcriptomics to determine a novel developmental trajectory for thyrotropes and potential novel regulators of thyrotrope populations., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
17. Targeting of a New Node in Lipid Metabolism as a Potential Treatment Strategy for ACC.
- Author
-
LaPensee CR and Hammer GD
- Subjects
- Humans, Mitotane, Lipid Metabolism, Lipolysis, Lipid Droplets metabolism, Adrenocortical Carcinoma metabolism, Adrenal Cortex Neoplasms metabolism
- Published
- 2023
- Full Text
- View/download PDF
18. Update on Biology and Genomics of Adrenocortical Carcinomas: Rationale for Emerging Therapies.
- Author
-
Lerario AM, Mohan DR, and Hammer GD
- Subjects
- Humans, Mitotane therapeutic use, Adrenal Glands, Genomics, Adrenocortical Carcinoma drug therapy, Adrenocortical Carcinoma genetics, Adrenal Cortex Neoplasms drug therapy, Adrenal Cortex Neoplasms genetics
- 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., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
19. American Association of Endocrine Surgeons Guidelines for Adrenalectomy: Executive Summary.
- Author
-
Yip L, Duh QY, Wachtel H, Jimenez C, Sturgeon C, Lee C, Velázquez-Fernández D, Berber E, Hammer GD, Bancos I, Lee JA, Marko J, Morris-Wiseman LF, Hughes MS, Livhits MJ, Han MA, Smith PW, Wilhelm S, Asa SL, Fahey TJ 3rd, McKenzie TJ, Strong VE, and Perrier ND
- Subjects
- Adrenalectomy methods, Cosyntropin, Glucocorticoids, Humans, Hydrocortisone, Adrenal Gland Neoplasms surgery, Pheochromocytoma surgery, Surgeons
- Abstract
Importance: Adrenalectomy is the definitive treatment for multiple adrenal abnormalities. Advances in technology and genomics and an improved understanding of adrenal pathophysiology have altered operative techniques and indications., Objective: To develop evidence-based recommendations to enhance the appropriate, safe, and effective approaches to adrenalectomy., Evidence Review: A multidisciplinary panel identified and investigated 7 categories of relevant clinical concern to practicing surgeons. Questions were structured in the framework Population, Intervention/Exposure, Comparison, and Outcome, and a guided review of medical literature from PubMed and/or Embase from 1980 to 2021 was performed. Recommendations were developed using Grading of Recommendations, Assessment, Development and Evaluation methodology and were discussed until consensus, and patient advocacy representation was included., Findings: Patients with an adrenal incidentaloma 1 cm or larger should undergo biochemical testing and further imaging characterization. Adrenal protocol computed tomography (CT) should be used to stratify malignancy risk and concern for pheochromocytoma. Routine scheduled follow-up of a nonfunctional adrenal nodule with benign imaging characteristics and unenhanced CT with Hounsfield units less than 10 is not suggested. When unilateral disease is present, laparoscopic adrenalectomy is recommended for patients with primary aldosteronism or autonomous cortisol secretion. Patients with clinical and radiographic findings consistent with adrenocortical carcinoma should be treated at high-volume multidisciplinary centers to optimize outcomes, including, when possible, a complete R0 resection without tumor disruption, which may require en bloc radical resection. Selective or nonselective α blockade can be used to safely prepare patients for surgical resection of paraganglioma/pheochromocytoma. Empirical perioperative glucocorticoid replacement therapy is indicated for patients with overt Cushing syndrome, but for patients with mild autonomous cortisol secretion, postoperative day 1 morning cortisol or cosyntropin stimulation testing can be used to determine the need for glucocorticoid replacement therapy. When patient and tumor variables are appropriate, we recommend minimally invasive adrenalectomy over open adrenalectomy because of improved perioperative morbidity. Minimally invasive adrenalectomy can be achieved either via a retroperitoneal or transperitoneal approach depending on surgeon expertise, as well as tumor and patient characteristics., Conclusions and Relevance: Twenty-six clinically relevant and evidence-based recommendations are provided to assist surgeons with perioperative adrenal care.
- Published
- 2022
- Full Text
- View/download PDF
20. Bones and adrenal organogenesis: how embryonic osteocalcin influences lifelong adrenal function.
- Author
-
Dumontet T and Hammer GD
- Subjects
- Animals, Glucocorticoids pharmacology, Osteocalcin genetics, Osteogenesis drug effects, Bone and Bones, Osteoblasts
- Abstract
Osteocalcin is a hormone produced in bones by osteoblasts during bone formation. Numerous studies have demonstrated that adrenal gland-derived glucocorticoids inhibit osteocalcin production, which can ultimately cause deleterious bones loss. This loss establishes a unidirectional endocrine relationship between the adrenal glands and bone, however, whether osteocalcin reciprocally regulates glucocorticoid secretion remains unclear. In this issue of the JCI, Yadav and colleagues address how bone-derived osteocalcin influences adrenal organogenesis and function. Using a large variety of animal models, the authors established that embryonic osteocalcin signaling, specifically through the GPR158 receptor, regulates postnatal adrenal steroid concentrations throughout life. This work has translational potential, and we await future investigations that determine whether modulating osteocalcin levels could promote endogenous adrenocortical function in adrenocortical hypoplasia and glucocorticoid deficiency.
- Published
- 2022
- Full Text
- View/download PDF
21. What Did We Learn from the Molecular Biology of Adrenal Cortical Neoplasia? From Histopathology to Translational Genomics.
- Author
-
Juhlin CC, Bertherat J, Giordano TJ, Hammer GD, Sasano H, and Mete O
- Subjects
- Adrenal Cortex Neoplasms diagnosis, Humans, Prognosis, Proteomics, Translational Research, Biomedical, Adrenal Cortex Neoplasms genetics, Adrenal Cortex Neoplasms pathology, Genomics, Molecular Biology
- Abstract
Approximately one-tenth of the general population exhibit adrenal cortical nodules, and the incidence has increased. Afflicted patients display a multifaceted symptomatology-sometimes with rather spectacular features. Given the general infrequency as well as the specific clinical, histological, and molecular considerations characterizing these lesions, adrenal cortical tumors should be investigated by endocrine pathologists in high-volume tertiary centers. Even so, to distinguish specific forms of benign adrenal cortical lesions as well as to pinpoint malignant cases with the highest risk of poor outcome is often challenging using conventional histology alone, and molecular genetics and translational biomarkers are therefore gaining increased attention as a possible discriminator in this context. In general, our understanding of adrenal cortical tumorigenesis has increased tremendously the last decade, not least due to the development of next-generation sequencing techniques. Comprehensive analyses have helped establish the link between benign aldosterone-producing adrenal cortical proliferations and ion channel mutations, as well as mutations in the protein kinase A (PKA) signaling pathway coupled to cortisol-producing adrenal cortical lesions. Moreover, molecular classifications of adrenal cortical tumors have facilitated the distinction of benign from malignant forms, as well as the prognostication of the individual patients with verified adrenal cortical carcinoma, enabling high-resolution diagnostics that is not entirely possible by histology alone. Therefore, combinations of histology, immunohistochemistry, and next-generation multi-omic analyses are all needed in an integrated fashion to properly distinguish malignancy in some cases. Despite significant progress made in the field, current clinical and pathological challenges include the preoperative distinction of non-metastatic low-grade adrenal cortical carcinoma confined to the adrenal gland, adoption of individualized therapeutic algorithms aligned with molecular and histopathologic risk stratification tools, and histological confirmation of functional adrenal cortical disease in the context of multifocal adrenal cortical proliferations. We herein review the histological, genetic, and epigenetic landscapes of benign and malignant adrenal cortical neoplasia from a modern surgical endocrine pathology perspective and highlight key mechanisms of value for diagnostic and prognostic purposes.
- Published
- 2021
- Full Text
- View/download PDF
22. β-catenin in adrenal zonation and disease.
- Author
-
Little DW 3rd, Dumontet T, LaPensee CR, and Hammer GD
- Subjects
- Animals, Humans, Ligands, Regeneration, Wnt Signaling Pathway, Adrenal Cortex metabolism, Adrenal Cortex pathology, Adrenal Gland Diseases metabolism, Adrenal Gland Diseases pathology, beta Catenin metabolism
- Abstract
The Wnt signaling pathway is a critical mediator of the development and maintenance of several tissues. The adrenal cortex is highly dependent upon Wnt/β-catenin signaling for proper zonation and endocrine function. Adrenocortical cells emerge in the peripheral capsule and subcapsular cortex of the gland as progenitor cells that centripetally differentiate into steroid hormone-producing cells of three functionally distinct concentric zones that respond robustly to various endocrine stimuli. Wnt/β-catenin signaling mediates adrenocortical progenitor cell fate and tissue renewal to maintain the gland throughout life. Aberrant Wnt/β-catenin signaling contributes to various adrenal disorders of steroid production and growth that range from hypofunction and hypoplasia to hyperfunction, hyperplasia, benign adrenocortical adenomas, and malignant adrenocortical carcinomas. Great strides have been made in defining the molecular underpinnings of adrenocortical homeostasis and disease, including the interplay between the capsule and cortex, critical components involved in maintaining the adrenocortical Wnt/β-catenin signaling gradient, and new targets in adrenal cancer. This review seeks to examine these and other recent advancements in understanding adrenocortical Wnt/β-catenin signaling and how this knowledge can inform therapeutic options for adrenal disease., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
23. Stem cell function and plasticity in the normal physiology of the adrenal cortex.
- Author
-
Hammer GD and Basham KJ
- Subjects
- Animals, Female, Humans, Male, Models, Biological, Sex Characteristics, Wnt Signaling Pathway, Adrenal Cortex cytology, Adrenal Cortex physiology, Cell Plasticity physiology, Stem Cells cytology, Stem Cells metabolism
- Abstract
The adrenal cortex functions to produce steroid hormones necessary for life. To maintain its functional capacity throughout life, the adrenal cortex must be continually replenished and rapidly repaired following injury. Moreover, the adrenal responds to endocrine-mediated organismal needs, which are highly dynamic and necessitate a precise steroidogenic response. To meet these diverse needs, the adrenal employs multiple cell populations with stem cell function. Here, we discuss the literature on adrenocortical stem cells using hematopoietic stem cells as a benchmark to examine the functional capacity of particular cell populations, including those located in the capsule and peripheral cortex. These populations are coordinately regulated by paracrine and endocrine signaling mechanisms, and display remarkable plasticity to adapt to different physiological and pathological conditions. Some populations also exhibit sex-specific activity, which contributes to highly divergent proliferation rates between sexes. Understanding mechanisms that govern adrenocortical renewal has broad implications for both regenerative medicine and cancer., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
24. Targeted RNAseq of Formalin-Fixed Paraffin-Embedded Tissue to Differentiate Among Benign and Malignant Adrenal Cortical Tumors.
- Author
-
Plaska SW, Liu CJ, Lim JS, Rege J, Bick NR, Lerario AM, Hammer GD, Giordano TJ, Else T, Tomlins SA, Rainey WE, and Udager AM
- Subjects
- Adrenal Cortex Neoplasms genetics, Adult, Aged, Aged, 80 and over, Cohort Studies, Diagnosis, Differential, Female, Follow-Up Studies, Formaldehyde chemistry, Gene Expression Regulation, Neoplastic, Humans, Male, Middle Aged, Prognosis, Adrenal Cortex Neoplasms classification, Adrenal Cortex Neoplasms diagnosis, Biomarkers, Tumor genetics, Paraffin Embedding methods, RNA-Seq methods, Transcriptome
- Abstract
Lack of routine fresh or frozen tissue is a barrier to widespread transcriptomic analysis of adrenal cortical tumors and an impediment to translational research in endocrinology and endocrine oncology. Our group has previously pioneered the use of targeted amplicon-based next-generation sequencing for archival formalin-fixed paraffin-embedded (FFPE) adrenal tissue specimens to characterize the spectrum of somatic mutations in various forms of primary aldosteronism. Herein, we developed and validated a novel 194-amplicon targeted next-generation RNA sequencing (RNAseq) assay for transcriptomic analysis of adrenal tumors using clinical-grade FFPE specimens. Targeted RNAseq-derived expression values for 27 adrenal cortical tumors, including aldosterone-producing adenomas (APA; n=8), cortisol-producing adenomas (CPA; n=11), and adrenal cortical carcinomas (ACC; n=8), highlighted known differentially-expressed genes (DEGs; i. e., CYP11B2 , IGF2 , etc.) and tumor type-specific transcriptional modules (i. e., high cell cycle/proliferation transcript expression in ACC, etc.), and a subset of DEGs was validated orthogonally using quantitative reverse transcription PCR (qRT-PCR). Finally, unsupervised hierarchical clustering using a subset of high-confidence DEGs revealed three discrete clusters representing APA, CPA, and ACC tumors with corresponding unique gene expression signatures, suggesting potential clinical utility for a transcriptomic-based approach to tumor classification. Overall, these data support the use of targeted amplicon-based RNAseq for comprehensive transcriptomic profiling of archival FFPE adrenal tumor material and indicate that this approach may facilitate important translational research opportunities for the study of these tumors., Competing Interests: The authors declare that they have no conflict of interest., (© Georg Thieme Verlag KG Stuttgart · New York.)
- Published
- 2020
- Full Text
- View/download PDF
25. New strategies for applying targeted therapies to adrenocortical carcinoma.
- Author
-
Mohan DR, Lerario AM, Finco I, and Hammer GD
- Abstract
Adrenocortical carcinoma (ACC) is a rare, aggressive, and frequently deadly cancer. Up to 75% of all patients will eventually develop metastatic disease, and our current medical therapies for ACC provide limited - if any - survival benefit. These statistics highlight a crucial need for novel approaches. Recent studies performing comprehensive molecular profiling on ACC have illuminated that ACC is comprised of three clinically distinct molecular subtypes, bearing differential regulation of cell cycle, epigenetics, Wnt/β-catenin signaling, PKA signaling, steroidogenesis and immune cell biology. Furthermore, these studies have spurred the development of molecular subtype-based biomarkers, contextualized outcomes of recent clinical trials, and advanced our understanding of the underlying biology of adrenocortical homeostasis and cancer. In this review, we describe these findings and their implications for new strategies to apply targeted therapies to ACC., Competing Interests: Conflict of Interest D.R. Mohan, A.M. Lerario, and G.D. Hammer are co-inventors on a provisional patent application describing compositions and methods for characterizing cancer, owned by The Regents of the University of Michigan. G.D. Hammer is founder and advisor for Millendo Therapeutics. G. D. Hammer is also founder of Vasaragen.
- Published
- 2019
- Full Text
- View/download PDF
26. Regulation of stem and progenitor cells in the adrenal cortex.
- Author
-
Finco I, Mohan DR, Hammer GD, and Lerario AM
- Abstract
The adrenal cortex is an endocrine organ comprised of three histological zones, the outermost zona glomerulosa, the intermediate zona fasciculata, and the innermost zona reticularis. High plasticity of the adrenal gland is supported by pools of stem and progenitor cells that are deployed to sustain physiological and homeostatic demands. In recent decades, exciting new discoveries elucidating the identity, function, and fate of these cell populations have emerged. In this review, we describe paracrine and endocrine signaling loops that are crucial for adrenal biology, focusing on recent studies unpacking the enigmatic nature of adrenal stem and progenitor cell populations., Competing Interests: Conflict of interest statement Nothing declared.
- Published
- 2019
- Full Text
- View/download PDF
27. Adjuvant Radiation Improves Recurrence-Free Survival and Overall Survival in Adrenocortical Carcinoma.
- Author
-
Gharzai LA, Green MD, Griffith KA, Else T, Mayo CS, Hesseltine E, Spratt DE, Ben-Josef E, Sabolch A, Miller BS, Worden F, Giordano TJ, Hammer GD, and Jolly S
- Subjects
- Adolescent, Adrenal Cortex Neoplasms pathology, Adrenal Cortex Neoplasms radiotherapy, Adrenocortical Carcinoma pathology, Adrenocortical Carcinoma radiotherapy, Adult, Aged, Case-Control Studies, Female, Follow-Up Studies, Humans, Male, Middle Aged, Neoplasm Recurrence, Local pathology, Neoplasm Recurrence, Local radiotherapy, Prognosis, Retrospective Studies, Survival Rate, Young Adult, Adrenal Cortex Neoplasms mortality, Adrenocortical Carcinoma mortality, Neoplasm Recurrence, Local mortality, Radiotherapy, Adjuvant mortality
- Abstract
Context: Adrenocortical carcinoma (ACC) is a rare malignancy with high rates of recurrence and poor prognosis. The role of radiotherapy (RT) in localized ACC has been controversial, and RT is not routinely offered., Objective: To evaluate the benefit of adjuvant RT on outcomes in ACC., Design: This is a retrospective propensity-matched analysis., Setting: All patients were seen through the University of Michigan's Endocrine Oncology program, and all those who underwent RT were treated at the University of Michigan., Participants: Of 424 patients with ACC, 78 were selected; 39 patients underwent adjuvant radiation., Intervention: Adjuvant RT to the tumor bed and adjacent lymph nodes., Main Outcomes Measures: Time to local failure, distant failure, or death., Results: Median follow-up time was 4.21 years (95% CI, 2.79 to 4.94). The median radiation dose was 55 Gy (range, 45 to 60). The 3-year overall survival estimate for patients improved from 48.6% for patients without RT (95% CI, 29.7 to 65.2) to 77.7% (95% CI, 56.3 to 89.5) with RT, with a hazard ratio (HR) of 3.59 (95% CI, 1.60 to 8.09; P = 0.002). RT improved local recurrence-free survival (RFS) from 34.2% (95% CI, 18.8 to 50.3) to 59.5% (95% CI, 39.0 to 75.0), with an HR of 2.67 (95% CI, 1.38 to 5.19; P = 0.0035). RT improved all RFS from 18.3% (95% CI, 6.7 to 34.3) to 46.7% (95% CI, 26.9 to 64.3), with an HR 2.59 (95% CI, 1.40 to 4.79; P = 0.0024)., Conclusions: In the largest single institution study to date, adjuvant RT after gross resection of ACC improved local RFS, all RFS, and overall survival in this propensity-matched analysis. Adjuvant RT should be considered a part of multidisciplinary management for patients with ACC., (Copyright © 2019 Endocrine Society.)
- Published
- 2019
- Full Text
- View/download PDF
28. Targeted Assessment of G0S2 Methylation Identifies a Rapidly Recurrent, Routinely Fatal Molecular Subtype of Adrenocortical Carcinoma.
- Author
-
Mohan DR, Lerario AM, Else T, Mukherjee B, Almeida MQ, Vinco M, Rege J, Mariani BMP, Zerbini MCN, Mendonca BB, Latronico AC, Marie SKN, Rainey WE, Giordano TJ, Fragoso MCBV, and Hammer GD
- Subjects
- Adrenal Cortex Neoplasms mortality, Adrenocortical Carcinoma mortality, Biomarkers, Tumor, Cell Line, Tumor, CpG Islands, Data Mining, Female, Gene Silencing, Genetic Loci, Humans, Male, Neoplasm Grading, Phenotype, Prognosis, Recurrence, Adrenal Cortex Neoplasms genetics, Adrenal Cortex Neoplasms pathology, Adrenocortical Carcinoma genetics, Adrenocortical Carcinoma pathology, Cell Cycle Proteins genetics, DNA Methylation
- Abstract
Purpose: Adrenocortical carcinoma (ACC) is a rare, aggressive malignancy with few therapies; however, patients with locoregional disease have variable outcomes. The Cancer Genome Atlas project on ACC (ACC-TCGA) identified that cancers of patients with homogeneously rapidly recurrent or fatal disease bear a unique CpG island hypermethylation phenotype, "CIMP-high." We sought to identify a biomarker that faithfully captures this subgroup. Experimental Design: We analyzed ACC-TCGA data to characterize differentially regulated biological processes, and identify a biomarker that is methylated and silenced exclusively in CIMP-high ACC. In an independent cohort of 114 adrenocortical tumors (80 treatment-naive primary ACC, 22 adrenocortical adenomas, and 12 non-naive/nonprimary ACC), we evaluated biomarker methylation by a restriction digest/qPCR-based approach, validated by targeted bisulfite sequencing. We evaluated expression of this biomarker and additional prognostic markers by qPCR., Results: We show that CIMP-high ACC is characterized by upregulation of cell cycle and DNA damage response programs, and identify that hypermethylation and silencing of G0S2 distinguishes this subgroup. We confirmed G0S2 hypermethylation and silencing is exclusive to 40% of ACC, and independently predicts shorter disease-free and overall survival (median 14 and 17 months, respectively). Finally, G0S2 methylation combined with validated molecular markers ( BUB1B-PINK1 ) stratifies ACC into three groups, with uniformly favorable, intermediate, and uniformly dismal outcomes., Conclusions: G0S2 hypermethylation is a hallmark of rapidly recurrent or fatal ACC, amenable to targeted assessment using routine molecular diagnostics. Assessing G0S2 methylation is straightforward, feasible for clinical decision-making, and will enable the direction of efficacious adjuvant therapies for patients with aggressive ACC., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
29. A ZNRF3-dependent Wnt/β-catenin signaling gradient is required for adrenal homeostasis.
- Author
-
Basham KJ, Rodriguez S, Turcu AF, Lerario AM, Logan CY, Rysztak MR, Gomez-Sanchez CE, Breault DT, Koo BK, Clevers H, Nusse R, Val P, and Hammer GD
- Subjects
- Adrenal Cortex cytology, Adrenal Cortex growth & development, Adrenal Cortex Diseases physiopathology, Animals, Cell Proliferation genetics, Female, Gene Knockout Techniques, Male, Mice, Models, Animal, Transcriptional Activation genetics, Ubiquitin-Protein Ligases genetics, Adrenal Cortex metabolism, Homeostasis genetics, Ubiquitin-Protein Ligases metabolism, Wnt Signaling Pathway physiology, beta Catenin metabolism
- Abstract
Spatiotemporal control of Wnt signaling is essential for the development and homeostasis of many tissues. The transmembrane E3 ubiquitin ligases ZNRF3 (zinc and ring finger 3) and RNF43 (ring finger protein 43) antagonize Wnt signaling by promoting degradation of frizzled receptors. ZNRF3 and RNF43 are frequently inactivated in human cancer, but the molecular and therapeutic implications remain unclear. Here, we demonstrate that adrenocortical-specific loss of ZNRF3, but not RNF43, results in adrenal hyperplasia that depends on Porcupine-mediated Wnt ligand secretion. Furthermore, we discovered a Wnt/β-catenin signaling gradient in the adrenal cortex that is disrupted upon loss of ZNRF3. Unlike β-catenin gain-of-function models, which induce high Wnt/β-catenin activation and expansion of the peripheral cortex, ZNRF3 loss triggers activation of moderate-level Wnt/β-catenin signaling that drives proliferative expansion of only the histologically and functionally distinct inner cortex. Genetically reducing β-catenin dosage significantly reverses the ZNRF3-deficient phenotype. Thus, homeostatic maintenance of the adrenal cortex is dependent on varying levels of Wnt/β-catenin activation, which is regulated by ZNRF3., (© 2019 Basham et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2019
- Full Text
- View/download PDF
30. Somatic mutations in adrenocortical carcinoma with primary aldosteronism or hyperreninemic hyperaldosteronism.
- Author
-
Mouat IC, Omata K, McDaniel AS, Hattangady NG, Talapatra D, Cani AK, Hovelson DH, Tomlins SA, Rainey WE, Hammer GD, Giordano TJ, and Else T
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Mutation, Retrospective Studies, Young Adult, Adrenocortical Carcinoma blood, High-Throughput Nucleotide Sequencing methods, Hyperaldosteronism etiology
- Abstract
Several somatic mutations specific to aldosterone-producing adenomas (APAs) have been described. A small proportion of adrenocortical carcinomas (ACCs) are associated with hyperaldosteronism, either primary aldosteronism or hyperreninemic hyperaldosteronism. However, it is unknown whether they harbor mutations of the same spectrum as APAs. The objective of this study is to describe the clinical phenotype and molecular genotype of ACCs with hyperaldosteronism, particularly the analysis for common APA-associated genetic changes. Patients were identified by retrospective chart review at a specialized referral center and by positive staining for CYP11B2 of tissue microarrays. Twenty-five patients with ACC and hyperaldosteronism were initially identified by retrospective chart review, and tissue for further analysis was available on 13 tumors. Seven patients were identified by positive staining for CYP11B2 in a tissue microarray, of which two were already identified in the initial chart review. Therefore, a total number of 18 patients with a diagnosis of ACC and features of either primary aldosteronism or hyperreninemic hyperaldosteronism were therefore included in the final study. Mutational status for a select list of oncogenes, tumor suppressor genes and genes known to carry mutations in APAs were analyzed by next-generation sequencing. Review of clinical data suggested autonomous aldosterone production in the majority of cases, while for some cases, hyperreninemic hyperaldosteronism was the more likely mechanism. The mutational landscape of ACCs associated with hyperaldosteronism was not different from ACCs with a different hormonal phenotype. None of the ACCs harbored mutations of known APA-associated genes, suggesting an alternative mechanism conferring aldosterone production.
- Published
- 2019
- Full Text
- View/download PDF
31. Longitudinal patterns of recurrence in patients with adrenocortical carcinoma.
- Author
-
Glenn JA, Else T, Hughes DT, Cohen MS, Jolly S, Giordano TJ, Worden FP, Gauger PG, Hammer GD, and Miller BS
- Subjects
- Adolescent, Adrenal Cortex Neoplasms pathology, Adrenal Cortex Neoplasms therapy, Adrenocortical Carcinoma pathology, Adrenocortical Carcinoma therapy, Adult, Aged, Aged, 80 and over, Female, Humans, Male, Metastasectomy statistics & numerical data, Michigan epidemiology, Middle Aged, Neoplasm Invasiveness, Neoplasm Metastasis, Neoplasm Recurrence, Local therapy, Radiotherapy, Adjuvant, Reoperation statistics & numerical data, Retrospective Studies, Time Factors, Young Adult, Adrenal Cortex Neoplasms mortality, Adrenocortical Carcinoma mortality, Neoplasm Recurrence, Local mortality
- Abstract
Background: Patterns and prognostic implications of recurrent adrenocortical carcinoma are poorly understood. In this study, we aim to describe temporal and spatial patterns of adrenocortical carcinoma recurrence., Methods: This is a retrospective review of 576 patients with adrenocortical carcinoma evaluated at a single institution. Clinicopathologic and follow-up data were collected longitudinally., Results: A total of 354 patients underwent resection of stage I-III adrenocortical carcinoma. We found that 249 (70%) patients developed disease recurrence. The median recurrence-free interval after primary resection was 11 months. The most common sites of initial recurrence were lung and tumor bed. The shortest time to recurrence was associated with lung or multiple site metastases. We found that 142 of 249 patients developed one or more additional sites of recurrence (median 5 months), most commonly involving the lungs. A total of 20 patients developed a third site of recurrence. We found that 100 patients underwent one or more reoperations or metastasectomies and 79 recurred again after reoperation. Same organ or site recurrence was common after reoperation (67%). Although lung metastases occurred early, recurrences to the peritoneal cavity or to multiple sites were associated with worse survival. Metastasectomy beyond three total operations did not improve overall survival., Conclusion: Survival varies according to site of recurrence and other clinicopathologic factors. Knowledge of patterns of recurrence may assist in anticipating disease course and lead to better informed selection of treatment., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
32. Isolation, Fixation, and Immunofluorescence Imaging of Mouse Adrenal Glands.
- Author
-
Finco I and Hammer GD
- Subjects
- Adrenal Glands pathology, Animals, Mice, Adrenal Glands diagnostic imaging
- Abstract
Immunofluorescence is a well-established technique for detection of antigens in tissues with the employment of fluorochrome-conjugated antibodies and has a broad spectrum of applications. Detection of antigens allows for characterization and identification of multiple cell types. Located above the kidneys and encapsulated by a layer of mesenchymal cells, the adrenal gland is an endocrine organ composed by two different tissues with different embryological origins, the mesonephric intermediate mesoderm-derived outer cortex and the neural crest-derived inner medulla. The adrenal cortex secretes steroids (i.e., mineralocorticoids, glucocorticoids, sex hormones), whereas the adrenal medulla produces catecholamines (i.e., adrenaline, noradrenaline). While conducting adrenal research, it is important to be able to distinguish unique cells with different functions. Here we provide a protocol developed in our laboratory that describes a series of sequential steps required for obtaining immunofluorescence staining to characterize the cell types of the adrenal gland. We focus first on the dissection of the mouse adrenal glands, the microscopic removal of periadrenal fat followed by the fixation, processing and paraffin embedding of the tissue. We then describe sectioning of the tissue blocks with a rotary microtome. Lastly, we detail a protocol for immunofluorescent staining of adrenal glands that we have developed to minimize both non-specific antibody binding and autofluorescence in order to achieve an optimal signal.
- Published
- 2018
- Full Text
- View/download PDF
33. Therapeutic Targets for Adrenocortical Carcinoma in the Genomics Era.
- Author
-
Mohan DR, Lerario AM, and Hammer GD
- Abstract
Adrenocortical carcinoma (ACC) is a rare and often fatal cancer, affecting ~1 person per million per year worldwide. Approximately 75% of patients with ACC eventually develop metastases and progress on the few available standard-of-care medical therapies, highlighting an incredible need for an improved understanding of the molecular biology of this disease. Although it has long been known that ACC is characterized by certain histological and genetic features ( e.g., high mitotic activity, chromosomal instability, and overexpression of IGF2 ), only in the last two decades of genomics has the molecular landscape of ACC been more thoroughly characterized. In this review, we describe the findings of historical genetics and recent genomics studies on ACC and discuss how underlying concepts emerging from these studies contribute to the current model of critical pathways for adrenocortical carcinogenesis. Integrative synthesis across these studies reveals that ACC consists of three distinct molecular subtypes with divergent clinical outcomes and implicates differential regulation of Wnt signaling, cell cycle, DNA methylation, immune biology, and steroidogenesis in ACC biology. These cellular programs are pharmacologically targetable and may enable the development of therapeutic strategies to improve outcomes for patients facing this devastating disease.
- Published
- 2018
- Full Text
- View/download PDF
34. Drug repurposing using high-throughput screening identifies a promising drug combination to treat adrenocortical carcinoma.
- Author
-
Lerario AM and Hammer GD
- Published
- 2018
- Full Text
- View/download PDF
35. Oncogenic Signaling Pathways in The Cancer Genome Atlas.
- Author
-
Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, Dimitriadoy S, Liu DL, Kantheti HS, Saghafinia S, Chakravarty D, Daian F, Gao Q, Bailey MH, Liang WW, Foltz SM, Shmulevich I, Ding L, Heins Z, Ochoa A, Gross B, Gao J, Zhang H, Kundra R, Kandoth C, Bahceci I, Dervishi L, Dogrusoz U, Zhou W, Shen H, Laird PW, Way GP, Greene CS, Liang H, Xiao Y, Wang C, Iavarone A, Berger AH, Bivona TG, Lazar AJ, Hammer GD, Giordano T, Kwong LN, McArthur G, Huang C, Tward AD, Frederick MJ, McCormick F, Meyerson M, Van Allen EM, Cherniack AD, Ciriello G, Sander C, and Schultz N
- Subjects
- Genes, Neoplasm, Humans, Neoplasms genetics, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Wnt Proteins genetics, Wnt Proteins metabolism, Databases, Genetic, Neoplasms pathology, Signal Transduction genetics
- Abstract
Genetic alterations in signaling pathways that control cell-cycle progression, apoptosis, and cell growth are common hallmarks of cancer, but the extent, mechanisms, and co-occurrence of alterations in these pathways differ between individual tumors and tumor types. Using mutations, copy-number changes, mRNA expression, gene fusions and DNA methylation in 9,125 tumors profiled by The Cancer Genome Atlas (TCGA), we analyzed the mechanisms and patterns of somatic alterations in ten canonical pathways: cell cycle, Hippo, Myc, Notch, Nrf2, PI-3-Kinase/Akt, RTK-RAS, TGFβ signaling, p53 and β-catenin/Wnt. We charted the detailed landscape of pathway alterations in 33 cancer types, stratified into 64 subtypes, and identified patterns of co-occurrence and mutual exclusivity. Eighty-nine percent of tumors had at least one driver alteration in these pathways, and 57% percent of tumors had at least one alteration potentially targetable by currently available drugs. Thirty percent of tumors had multiple targetable alterations, indicating opportunities for combination therapy., (Copyright © 2018. Published by Elsevier Inc.)
- Published
- 2018
- Full Text
- View/download PDF
36. Sonic Hedgehog and WNT Signaling Promote Adrenal Gland Regeneration in Male Mice.
- Author
-
Finco I, Lerario AM, and Hammer GD
- Subjects
- Adrenal Cortex drug effects, Adrenal Cortex metabolism, Adrenal Glands drug effects, Animals, Female, Glucocorticoids pharmacology, Hedgehog Proteins genetics, Male, Mice, Mice, Inbred C57BL, Regeneration drug effects, Signal Transduction drug effects, Stem Cells drug effects, Stem Cells metabolism, Wnt4 Protein genetics, Zinc Finger Protein GLI1 genetics, Zinc Finger Protein GLI1 metabolism, Zona Glomerulosa drug effects, Zona Glomerulosa metabolism, Adrenal Glands physiology, Hedgehog Proteins metabolism, Wnt4 Protein metabolism
- 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., (Copyright © 2018 Endocrine Society.)
- Published
- 2018
- Full Text
- View/download PDF
37. Timing of adrenal regression controlled by synergistic interaction between Sf1 SUMOylation and Dax1.
- Author
-
Xing Y, Morohashi KI, Ingraham HA, and Hammer GD
- Subjects
- Animals, DAX-1 Orphan Nuclear Receptor genetics, Female, HEK293 Cells, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Protein Processing, Post-Translational, Real-Time Polymerase Chain Reaction, Steroidogenic Factor 1 genetics, Sumoylation, Transcription, Genetic, Adrenal Cortex embryology, DAX-1 Orphan Nuclear Receptor physiology, Gene Expression Regulation, Developmental, Steroidogenic Factor 1 physiology
- Abstract
The nuclear receptor steroidogenic factor 1 (Sf1, Nr5a1, Ad4bp) is crucial for formation, development and function of steroidogenic tissues. A fetal adrenal enhancer (FAdE) in the Sf1 gene was previously identified to direct Sf1 expression exclusively in the fetal adrenal cortex and is bound by both Sf1 and Dax1. Here, we have examined the function of Sf1 SUMOylation and its interaction with Dax1 on FAdE function . A diffused prolonged pattern of FAdE expression and delayed regression of the postnatal fetal cortex (X-zone) were detected in both the SUMOylation-deficient- Sf1
2KR/2KR and Dax1 knockout mouse lines, with FAdE expression/activity retained in the postnatal 20αHSD-positive postnatal X-zone cells. In vitro studies indicated that Sf1 SUMOylation, although not directly influencing DNA binding, actually increased binding of Dax1 to Sf1 to further enhance transcriptional repression of FAdE. Taken together, these studies define a crucial repressor function of Sf1 SUMOylation and Dax1 in the physiological cessation of FAdE-mediated Sf1 expression and the resultant regression of the postnatal fetal cortex (X-zone)., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2017. Published by The Company of Biologists Ltd.)- Published
- 2017
- Full Text
- View/download PDF
38. Cell signaling pathways in the adrenal cortex: Links to stem/progenitor biology and neoplasia.
- Author
-
Penny MK, Finco I, and Hammer GD
- Subjects
- Adrenal Cortex cytology, Adrenal Cortex pathology, Hedgehog Proteins metabolism, Humans, Stem Cells cytology, Wnt Signaling Pathway, Adrenal Cortex metabolism, Adrenal Cortex Neoplasms metabolism, Stem Cells metabolism
- Abstract
The adrenal cortex is a dynamic tissue responsible for the synthesis of steroid hormones, including mineralocorticoids, glucocorticoids, and androgens in humans. Advances have been made in understanding the role of adrenocortical stem/progenitor cell populations in cortex homeostasis and self-renewal. Recently, large molecular profiling studies of adrenocortical carcinoma (ACC) have given insights into proteins and signaling pathways involved in normal tissue homeostasis that become dysregulated in cancer. These data provide an impetus to examine the cellular pathways implicated in adrenocortical disease and study connections, or lack thereof, between adrenal homeostasis and tumorigenesis, with a particular focus on stem and progenitor cell pathways. In this review, we discuss evidence for stem/progenitor cells in the adrenal cortex, proteins and signaling pathways that may regulate these cells, and the role these proteins play in pathologic and neoplastic conditions. In turn, we also examine common perturbations in adrenocortical tumors (ACT) and how these proteins and pathways may be involved in adrenal homeostasis., (Copyright © 2016 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
39. Molecular Mechanisms of Stem/Progenitor Cell Maintenance in the Adrenal Cortex.
- Author
-
Lerario AM, Finco I, LaPensee C, and Hammer GD
- 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.
- Published
- 2017
- Full Text
- View/download PDF
40. Comprehensive Pan-Genomic Characterization of Adrenocortical Carcinoma.
- Author
-
Zheng S, Cherniack AD, Dewal N, Moffitt RA, Danilova L, Murray BA, Lerario AM, Else T, Knijnenburg TA, Ciriello G, Kim S, Assie G, Morozova O, Akbani R, Shih J, Hoadley KA, Choueiri TK, Waldmann J, Mete O, Robertson AG, Wu HT, Raphael BJ, Shao L, Meyerson M, Demeure MJ, Beuschlein F, Gill AJ, Sidhu SB, Almeida MQ, Fragoso MCBV, Cope LM, Kebebew E, Habra MA, Whitsett TG, Bussey KJ, Rainey WE, Asa SL, Bertherat J, Fassnacht M, Wheeler DA, Hammer GD, Giordano TJ, and Verhaak RGW
- Published
- 2016
- Full Text
- View/download PDF
41. Double adrenocortical adenomas harboring independent KCNJ5 and PRKACA somatic mutations.
- Author
-
Nanba K, Omata K, Tomlins SA, Giordano TJ, Hammer GD, Rainey WE, and Else T
- Subjects
- Adrenal Cortex Neoplasms blood, Adrenal Cortex Neoplasms pathology, Adrenal Cortex Neoplasms surgery, Adrenocortical Adenoma blood, Adrenocortical Adenoma pathology, Adrenocortical Adenoma surgery, Aldosterone blood, Cushing Syndrome blood, Cushing Syndrome genetics, Cushing Syndrome pathology, Cushing Syndrome surgery, Female, Humans, Hydrocortisone blood, Hyperaldosteronism blood, Hyperaldosteronism genetics, Hyperaldosteronism pathology, Hyperaldosteronism surgery, Middle Aged, Neoplasms, Multiple Primary blood, Neoplasms, Multiple Primary pathology, Neoplasms, Multiple Primary surgery, Treatment Outcome, Adrenal Cortex Neoplasms genetics, Adrenocortical Adenoma genetics, Cyclic AMP-Dependent Protein Kinase Catalytic Subunits genetics, G Protein-Coupled Inwardly-Rectifying Potassium Channels genetics, Mutation, Neoplasms, Multiple Primary genetics
- Abstract
Objective: Co-secretion of cortisol and aldosterone can be observed in adrenal adenomas. The aim of this study was to investigate the molecular characteristics of a co-existing aldosterone- and a cortisol-producing adenoma (CPA) in the same patient., Design and Methods: Two different adenomas within the same adrenal gland from a 49-year-old female patient with primary aldosteronism (PA) and Cushing's syndrome (CS) were studied. Multiple formalin-fixed paraffin-embedded tumor blocks were used for the analysis. Immunohistochemistry (IHC) was performed using a specific antibody against aldosterone synthase (CYP11B2). DNA and RNA were isolated separately from CYP11B2-positive and -negative tumor regions based on CYP11B2 IHC results., Results: CYP11B2 IHC clearly demonstrated that three pieces from one adenoma were positive for CYP11B2 and the remaining three from the other adenoma were negative for CYP11B2. In quantitative real-time RT-PCR, CYP11B2 mRNA was upregulated in CYP11B2-positive tumor specimens (219-fold vs CYP11B2-negative tumor specimens). Targeted next-generation sequencing (NGS) detected novel KCNJ5 gene mutations (p.T148I/T149S, present in the same reads) and a PRKACA gene hotspot mutation (p.L206R) in the CYP11B2-positive and -negative tumors, respectively. Sanger sequencing of DNA from each tumor specimen (CYP11B2-positive tumor, n=3; CYP11B2-negative tumor, n=3) showed concordant results with targeted NGS., Conclusion: Our findings illustrate the co-existence of two different adrenocortical adenomas causing the concurrent diagnosis of PA and CS in the same patient. Molecular analysis was able to demonstrate that the two diseases resulted from independent somatic mutations seen in double adrenocortical adenomas., (© 2016 European Society of Endocrinology.)
- Published
- 2016
- Full Text
- View/download PDF
42. ATR-101, a Selective and Potent Inhibitor of Acyl-CoA Acyltransferase 1, Induces Apoptosis in H295R Adrenocortical Cells and in the Adrenal Cortex of Dogs.
- Author
-
LaPensee CR, Mann JE, Rainey WE, Crudo V, Hunt SW 3rd, and Hammer GD
- Subjects
- Adrenal Cortex metabolism, Adrenal Cortex Neoplasms metabolism, Adrenal Cortex Neoplasms pathology, Adrenocortical Carcinoma metabolism, Adrenocortical Carcinoma pathology, Animals, Caspase 3 metabolism, Cell Line, Tumor, Dogs, Humans, Membrane Potential, Mitochondrial drug effects, Sterol O-Acyltransferase metabolism, Adrenal Cortex drug effects, Apoptosis drug effects, Phenylurea Compounds pharmacology, Sterol O-Acyltransferase antagonists & inhibitors
- Abstract
ATR-101 is a novel, oral drug candidate currently in development for the treatment of adrenocortical cancer. ATR-101 is a selective and potent inhibitor of acyl-coenzyme A:cholesterol O-acyltransferase 1 (ACAT1), an enzyme located in the endoplasmic reticulum (ER) membrane that catalyzes esterification of intracellular free cholesterol (FC). We aimed to identify mechanisms by which ATR-101 induces adrenocortical cell death. In H295R human adrenocortical carcinoma cells, ATR-101 decreases the formation of cholesteryl esters and increases FC levels, demonstrating potent inhibition of ACAT1 activity. Caspase-3/7 levels and terminal deoxynucleotidyl transferase 2'-deoxyuridine 5'-triphosphate nick end labeled-positive cells are increased by ATR-101 treatment, indicating activation of apoptosis. Exogenous cholesterol markedly potentiates the activity of ATR-101, suggesting that excess FC that cannot be adequately esterified increases caspase-3/7 activation and subsequent cell death. Inhibition of calcium release from the ER or the subsequent uptake of calcium by mitochondria reverses apoptosis induced by ATR-101. ATR-101 also activates multiple components of the unfolded protein response, an indicator of ER stress. Targeted knockdown of ACAT1 in an adrenocortical cell line mimicked the effects of ATR-101, suggesting that ACAT1 mediates the cytotoxic effects of ATR-101. Finally, in vivo treatment of dogs with ATR-101 decreased adrenocortical steroid production and induced cellular apoptosis that was restricted to the adrenal cortex. Together, these studies demonstrate that inhibition of ACAT1 by ATR-101 increases FC, resulting in dysregulation of ER calcium stores that result in ER stress, the unfolded protein response, and ultimately apoptosis.
- Published
- 2016
- Full Text
- View/download PDF
43. Molecular Heterogeneity in Aldosterone-Producing Adenomas.
- Author
-
Nanba K, Chen AX, Omata K, Vinco M, Giordano TJ, Else T, Hammer GD, Tomlins SA, and Rainey WE
- Subjects
- Adrenal Glands pathology, Adrenalectomy, Adrenocortical Adenoma pathology, DNA analysis, DNA chemistry, Gene Expression, Genetic Heterogeneity, Humans, Hyperaldosteronism pathology, Immunohistochemistry, Mutation, Sequence Analysis, DNA, Adrenocortical Adenoma chemistry, Adrenocortical Adenoma genetics, Aldosterone biosynthesis, Cytochrome P-450 CYP11B2 genetics, Hyperaldosteronism genetics
- Abstract
Context: The use of next-generation sequencing has resulted in the identification of recurrent somatic mutations underlying primary aldosteronism (PA). However, significant gaps remain in our understanding of the relationship between tumor aldosterone synthase (CYP11B2) expression and somatic mutation status., Objective: The objective of the study was to investigate tumor CYP11B2 expression and somatic aldosterone-driver gene mutation heterogeneity., Methods: Fifty-one adrenals from 51 PA patients were studied. Immunohistochemistry for CYP11B2 was performed. Aldosterone-producing adenomas with intratumor CYP11B2 heterogeneity were analyzed for mutation status using targeted next-generation sequencing. DNA was isolated from CYP11B2-positive, CYP11B2-negative, and adjacent normal areas from formalin-fixed, paraffin-embedded sections., Results: Of 51 adrenals, seven (14 %) showed distinct heterogeneity in CYP11B2 by immunohistochemistry, including six adenomas with intratumor heterogeneity and one multinodular hyperplastic adrenal with both CYP11B2-positive and -negative nodules. Of the six adrenocortical adenomas with CYP11B2 heterogeneity, three had aldosterone-regulating mutations (CACNA1D p.F747C, KCNJ5 p.L168R, ATP1A1 p.L104R) only in CYP11B2-positive regions, and one had two different mutations localized to two histologically distinct CYP11B2-positive regions (ATP2B3 p.L424_V425del, KCNJ5 p.G151R). Lastly, one adrenal with multiple CYP11B2-expressing nodules showed different mutations in each (CACNA1D p.F747V and ATP1A1 p.L104R), and no mutations were identified in CYP11B2-negative nodule or adjacent normal adrenal., Conclusions: Adrenal tumors in patients with PA can demonstrate clear heterogeneity in CYP11B2 expression and somatic mutations in driver genes for aldosterone production. These findings suggest that aldosterone-producing adenoma tumorigenesis can occur within preexisting nodules through the acquisition of somatic mutations that drive aldosterone production.
- Published
- 2016
- Full Text
- View/download PDF
44. Mouse models of adrenocortical tumors.
- Author
-
Basham KJ, Hung HA, Lerario AM, and Hammer GD
- Subjects
- Adrenal Cortex Neoplasms metabolism, Animals, Humans, Insulin-Like Growth Factor II genetics, Mice, Tumor Suppressor Protein p53 genetics, Wnt Signaling Pathway, Adrenal Cortex Neoplasms genetics, Adrenal Cortex Neoplasms pathology, Disease Models, Animal
- Abstract
The molecular basis of the organogenesis, homeostasis, and tumorigenesis of the adrenal cortex has been the subject of intense study for many decades. Specifically, characterization of tumor predisposition syndromes with adrenocortical manifestations and molecular profiling of sporadic adrenocortical tumors have led to the discovery of key molecular pathways that promote pathological adrenal growth. However, given the observational nature of such studies, several important questions regarding the molecular pathogenesis of adrenocortical tumors have remained. This review will summarize naturally occurring and genetically engineered mouse models that have provided novel tools to explore the molecular and cellular underpinnings of adrenocortical tumors. New paradigms of cancer initiation, maintenance, and progression that have emerged from this work will be discussed., (Copyright © 2015 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
45. 5th International ACC Symposium: Introduction.
- Author
-
Hammer GD
- Subjects
- Adrenal Cortex Neoplasms pathology, Adrenal Cortex Neoplasms therapy, Adrenal Glands pathology, Humans, Adrenal Cortex Neoplasms diagnosis
- Published
- 2016
- Full Text
- View/download PDF
46. Diagnosis and Treatment of Primary Adrenal Insufficiency: An Endocrine Society Clinical Practice Guideline.
- Author
-
Bornstein SR, Allolio B, Arlt W, Barthel A, Don-Wauchope A, Hammer GD, Husebye ES, Merke DP, Murad MH, Stratakis CA, and Torpy DJ
- Subjects
- Evidence-Based Medicine, Female, Hormone Replacement Therapy methods, Humans, Pregnancy, Societies, Medical, Adrenal Insufficiency diagnosis, Adrenal Insufficiency therapy
- Abstract
Objective: This clinical practice guideline addresses the diagnosis and treatment of primary adrenal insufficiency., Participants: The Task Force included a chair, selected by The Clinical Guidelines Subcommittee of the Endocrine Society, eight additional clinicians experienced with the disease, a methodologist, and a medical writer. The co-sponsoring associations (European Society of Endocrinology and the American Association for Clinical Chemistry) had participating members. The Task Force received no corporate funding or remuneration in connection with this review., Evidence: This evidence-based guideline was developed using the Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) system to determine the strength of recommendations and the quality of evidence., Consensus Process: The evidence used to formulate recommendations was derived from two commissioned systematic reviews as well as other published systematic reviews and studies identified by the Task Force. The guideline was reviewed and approved sequentially by the Endocrine Society's Clinical Guidelines Subcommittee and Clinical Affairs Core Committee, members responding to a web posting, and the Endocrine Society Council. At each stage, the Task Force incorporated changes in response to written comments., Conclusions: We recommend diagnostic tests for the exclusion of primary adrenal insufficiency in all patients with indicative clinical symptoms or signs. In particular, we suggest a low diagnostic (and therapeutic) threshold in acutely ill patients, as well as in patients with predisposing factors. This is also recommended for pregnant women with unexplained persistent nausea, fatigue, and hypotension. We recommend a short corticotropin test (250 μg) as the "gold standard" diagnostic tool to establish the diagnosis. If a short corticotropin test is not possible in the first instance, we recommend an initial screening procedure comprising the measurement of morning plasma ACTH and cortisol levels. Diagnosis of the underlying cause should include a validated assay of autoantibodies against 21-hydroxylase. In autoantibody-negative individuals, other causes should be sought. We recommend once-daily fludrocortisone (median, 0.1 mg) and hydrocortisone (15-25 mg/d) or cortisone acetate replacement (20-35 mg/d) applied in two to three daily doses in adults. In children, hydrocortisone (∼8 mg/m(2)/d) is recommended. Patients should be educated about stress dosing and equipped with a steroid card and glucocorticoid preparation for parenteral emergency administration. Follow-up should aim at monitoring appropriate dosing of corticosteroids and associated autoimmune diseases, particularly autoimmune thyroid disease.
- Published
- 2016
- Full Text
- View/download PDF
47. Nutritional conditions regulate transcriptional activity of SF-1 by controlling sumoylation and ubiquitination.
- Author
-
Lee J, Yang DJ, Lee S, Hammer GD, Kim KW, and Elmquist JK
- Subjects
- Animals, Cell Line, Humans, Insulin metabolism, Insulin pharmacology, Models, Biological, Protein Stability, Proteolysis, Signal Transduction, Gene Expression Regulation drug effects, Steroidogenic Factor 1 metabolism, Sumoylation drug effects, Transcriptional Activation, Ubiquitination drug effects
- Abstract
Steroidogenic factor 1 (SF-1) is a transcription factor expressed in the ventral medial nucleus of the hypothalamus that regulates energy homeostasis. However, the molecular mechanisms of SF-1 in the control of energy balance are largely unknown. Here, we show that nutritional conditions, such as the presence or absence of serum, affect SF-1 action. Serum starvation significantly decreased hypothalamic SF-1 levels by promoting ubiquitin-dependent degradation, and sumoylation was required for this process. SF-1 transcriptional activity was also differentially regulated by nutritional status. Under normal conditions, the transcriptional activity of hypothalamic SF-1 was activated by SUMO, but this was attenuated during starvation. Taken together, these results indicate that sumoylation and ubiquitination play crucial roles in the regulation of SF-1 function and that these effects are dependent on nutritional conditions, further supporting the importance of SF-1 in the control of energy homeostasis.
- Published
- 2016
- Full Text
- View/download PDF
48. Radiographic Characteristics of Adrenal Masses Preceding the Diagnosis of Adrenocortical Cancer.
- Author
-
Nogueira TM, Lirov R, Caoili EM, Lerario AM, Miller BS, Fragoso MC, Dunnick NR, Hammer GD, and Else T
- Subjects
- Adrenal Cortex Neoplasms pathology, Adult, Female, Humans, Male, Middle Aged, Prognosis, Retrospective Studies, Tomography, X-Ray Computed, Adrenal Cortex Neoplasms diagnosis, Adrenal Glands pathology
- Abstract
Incidentally discovered adrenal masses are common and the clinical evaluation and surveillance aims to diagnose hormone excess and malignancy. Adrenocortical cancer (ACC) is a very rare malignancy. This study aims to define the imaging characteristics of adrenal tumors preceding the diagnosis of ACC. Patients with prior (>5 months) adrenal tumors (<6 cm) subsequently diagnosed with ACC were identified in a large registry at a tertiary referral center. Retrospective chart and image review for patient characteristics and initial, interval, and diagnostic imaging characteristics (size, homogeneity, borders, density, growth rate, etc.) was conducted. Twenty patients with a diagnosis of ACC and a prior adrenal tumor were identified among 422 patients with ACC. Of these, 17 patients were initially imaged with CT and 3 with MR. Only 2 of the 20 patients had initial imaging characteristics suggestive of a benign lesion. Of initial tumors, 25% were <2 cm in size. Surveillance led to the diagnosis of ACC within 24 months in 50% of patients. The growth pattern was variable with some lesions showing long-term stability (up to 8 years) in size. In conclusion, antecedent lesions in patients with a diagnosis of ACC are often indeterminate by imaging criteria and can be small. Surveillance over 2 years detected only 50% of ACCs. Current practice and guidelines are insufficient in diagnosing ACCs. Given the rarity of ACC, the increased risk and health care costs of additional evaluation may not be warranted.
- Published
- 2015
- Full Text
- View/download PDF
49. Adjuvant radiation therapy improves local control after surgical resection in patients with localized adrenocortical carcinoma.
- Author
-
Sabolch A, Else T, Griffith KA, Ben-Josef E, Williams A, Miller BS, Worden F, Hammer GD, and Jolly S
- Subjects
- Adrenal Cortex Neoplasms mortality, Adrenal Cortex Neoplasms pathology, Adrenal Cortex Neoplasms surgery, Adrenocortical Carcinoma mortality, Adrenocortical Carcinoma pathology, Adrenocortical Carcinoma surgery, Adult, Aged, Cohort Studies, Female, Humans, Male, Michigan, Middle Aged, Neoplasm Staging, Radiotherapy Dosage, Radiotherapy, Adjuvant methods, Radiotherapy, Adjuvant mortality, Retrospective Studies, Universities, Young Adult, Adrenal Cortex Neoplasms radiotherapy, Adrenocortical Carcinoma radiotherapy
- Abstract
Purpose: Adrenocortical carcinoma (ACC) is a rare malignancy known for high rates of local recurrence, though the benefit of postoperative radiation therapy (RT) has not been established. In this study of grossly resected ACC, we compare local control of patients treated with surgery followed by adjuvant RT to a matched cohort treated with surgery alone., Methods and Materials: We retrospectively identified patients with localized disease who underwent R0 or R1 resection followed by adjuvant RT. Only patients treated with RT at our institution were included. Matching to surgical controls was on the basis of stage, surgical margin status, tumor grade, and adjuvant mitotane., Results: From 1991 to 2011, 360 ACC patients were evaluated for ACC at the University of Michigan (Ann Arbor, MI). Twenty patients with localized disease received postoperative adjuvant RT. These were matched to 20 controls. There were no statistically significant differences between the groups with regard to stage, margins, grade, or mitotane. Median RT dose was 55 Gy (range, 45-60 Gy). Median follow-up was 34 months. Local recurrence occurred in 1 patient treated with RT, compared with 12 patients not treated with RT (P=.0005; hazard ratio [HR] 12.59; 95% confidence interval [CI] 1.62-97.88). However, recurrence-free survival was no different between the groups (P=.17; HR 1.52; 95% CI 0.67-3.45). Overall survival was also not significantly different (P=.13; HR 1.97; 95% CI 0.57-6.77), with 4 deaths in the RT group compared with 9 in the control group., Conclusions: Postoperative RT significantly improved local control compared with the use of surgery alone in this case-matched cohort analysis of grossly resected ACC patients. Although this retrospective series represents the largest study to date on adjuvant RT for ACC, its findings need to be prospectively confirmed., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
50. Development of adrenal cortex zonation.
- Author
-
Xing Y, Lerario AM, Rainey W, and Hammer GD
- Subjects
- Adrenal Cortex growth & development, Adrenal Cortex metabolism, Androgens metabolism, Glucocorticoids metabolism, Hedgehog Proteins metabolism, Humans, Mineralocorticoids metabolism, Puberty metabolism, Zona Fasciculata embryology, Zona Fasciculata growth & development, Zona Fasciculata metabolism, Zona Glomerulosa embryology, Zona Glomerulosa growth & development, Zona Glomerulosa metabolism, Zona Reticularis embryology, Zona Reticularis growth & development, Zona Reticularis metabolism, Adrenal Cortex embryology, Cell Differentiation, Stem Cells metabolism
- Abstract
The human adult adrenal cortex is composed of the zona glomerulosa (zG), zona fasciculata (zF), and zona reticularis (zR), which are responsible for production of mineralocorticoids, glucocorticoids, and adrenal androgens, respectively. The final completion of cortical zonation in humans does not occur until puberty with the establishment of the zR and its production of adrenal androgens; a process called adrenarche. The maintenance of the adrenal cortex involves the centripetal displacement and differentiation of peripheral Sonic hedgehog-positive progenitors cells into zG cells that later transition to zF cells and subsequently zR cells., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.