1
|
Bizzi MF, Drummond JB, Pinheiro SVB, Paulino E, Araújo SA, Soares BS, Giannetti AV, Schweizer JRDOL, Barry S, Korbonits M, Ribeiro-Oliveira A. Activated AMP-protein kinase (pAMPK) is overexpressed in human somatotroph pituitary adenomas. Mol Cell Endocrinol 2024; 592:112318. [PMID: 38908427 DOI: 10.1016/j.mce.2024.112318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
INTRODUCTION AMPK (AMP-activated protein kinase) is an enzyme that acts as a metabolic sensor and regulates multiple pathways via phosphorylating proteins in metabolic and proliferative pathways. The aim of this work was to study the activated cellular AMPK (phosphorylated-AMPK at Thr172, pAMPK) levels in pituitary tumor samples from patients with sporadic and familial acromegaly, as well as in samples from normal human pituitary gland. METHODS We studied pituitary adenoma tissue from patients with sporadic somatotroph adenomas, familial acromegaly with heterozygote germline variants in the aryl hydrocarbon receptor interacting protein (AIP) gene (p.Q164*, p.R304* and p.F269_H275dup) and autopsy from normal pituitary glands without structural alterations. RESULTS Cellular levels of pAMPK were significantly higher in patients with sporadic acromegaly compared to normal pituitary glands (p < 0.0001). Tissues samples from patients with germline AIP mutations also showed higher cellular levels of pAMPK compared to normal pituitary glands. We did not observe a significant difference in cellular levels of pAMPK according to the cytokeratin (CAM5.2) pattern (sparsely or densely granulated) for tumor samples of sporadic acromegaly. CONCLUSION Our data show, for the first time in human cells, an increase of cellular levels of pAMPK in sporadic somatotropinomas, regardless of cytokeratin pattern, as well as in GH-secreting adenomas from patients with germline AIP mutations.
Collapse
Affiliation(s)
- Mariana Ferreira Bizzi
- Departments of Internal Medicine of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Juliana Beaudette Drummond
- Departments of Internal Medicine of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Sergio Veloso Brant Pinheiro
- Departments of Pediatrics of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Eduardo Paulino
- Departments of Pathology of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Stanley Almeida Araújo
- Departments of Pathology of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Beatriz Santana Soares
- Departments of Internal Medicine of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Alexandre V Giannetti
- Departments of Surgery of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | | | - Sayka Barry
- Centre for Endocrinology of Queen Mary University of London, London, EC1M 6BQ, UK
| | - Márta Korbonits
- Centre for Endocrinology of Queen Mary University of London, London, EC1M 6BQ, UK
| | - Antonio Ribeiro-Oliveira
- Departments of Internal Medicine of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil.
| |
Collapse
|
2
|
Vella M, Manfield IW, Seychell BC, Trinh CH, Rambo R, Nasir Khan G, Vassallo J, Hunter T, Hunter GJ. Mutations in the N-domain of aryl hydrocarbon receptor interacting protein affect interactions with heat shock protein 90β and phosphodiesterase 4A5. Biochimie 2024:S0300-9084(24)00215-3. [PMID: 39299536 DOI: 10.1016/j.biochi.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
The aryl hydrocarbon receptor interacting protein (AIP) is a cytoplasmic molecular co-chaperone and tumour suppressor that assists in protein stability and complex formation involving the aryl hydrocarbon receptor. Germline mutations in the AIP gene predispose to pituitary tumourigenesis with patients exhibiting an aggressive clinical phenotype. Full length AIP proteins harbouring N-domain mutations (R9Q, R16H, V49 M and K103R) were purified from E.coli utilizing a methodology that maintained structural integrity and monomeric stability. Mutations did not significantly affect the thermal stability of the protein and caused no overall disruptive effect in the protein structure. The mutations studied lowered the binding affinity of AIP towards two of its binding partners; heat shock protein 90β and phosphodiesterase 4A5 (PDE4A5). The inhibition of phosphodiesterase activity by AIP was also greatly reduced by all mutants. While previously published data has mainly concentrated on the tetratricopeptide repeats of the C-domain of AIP, we present clear evidence that AIP N-domain mutations play a significant role in two protein:protein interactions with partner proteins. The complex interactome of AIP suggests that any observable change in one or more of its binding partners cannot be disregarded as it may have repercussions on other biochemical pathways.
Collapse
Affiliation(s)
- Marita Vella
- Department of Physiology & Biochemistry, Faculty of Medicine & Surgery, University of Malta, Msida, MSD2080, Malta
| | - Iain W Manfield
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Brandon C Seychell
- Department of Physiology & Biochemistry, Faculty of Medicine & Surgery, University of Malta, Msida, MSD2080, Malta
| | - Chi H Trinh
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Robert Rambo
- Soft Condensed Matter Group, Diamond Light Source Ltd, Diamond House, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, UK
| | - G Nasir Khan
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Josanne Vassallo
- Department of Medicine, Faculty of Medicine & Surgery, University of Malta, Msida, MSD2080, Malta
| | - Thérèse Hunter
- Department of Physiology & Biochemistry, Faculty of Medicine & Surgery, University of Malta, Msida, MSD2080, Malta
| | - Gary J Hunter
- Department of Physiology & Biochemistry, Faculty of Medicine & Surgery, University of Malta, Msida, MSD2080, Malta.
| |
Collapse
|
3
|
Shen AL, Moran SM, Glover EN, Lin BC, Carney PR, Bradfield CA. Familial isolated pituitary adenoma is independent of Ahr genotype in a novel mouse model of disease. Heliyon 2024; 10:e28231. [PMID: 38590848 PMCID: PMC10999881 DOI: 10.1016/j.heliyon.2024.e28231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
Human familial isolated pituitary adenoma (FIPA) has been linked to germline heterozygous mutations in the gene encoding the aryl hydrocarbon receptor-interacting protein (AIP, also known as ARA9, XAP2, FKBP16, or FKBP37). To investigate the hypothesis that AIP is a pituitary adenoma tumor suppressor via its role in aryl hydrocarbon receptor (AHR) signaling, we have compared the pituitary phenotype of our global null Aip (AipΔC) mouse model with that of a conditional null Aip model (Aipfx/fx) carrying the same deletion, as well as pituitary phenotypes of Ahr global null and Arnt conditional null animals. We demonstrate that germline AipΔC heterozygosity results in a high incidence of pituitary tumors in both sexes, primarily somatotropinomas, at 16 months of age. Biallelic deletion of Aip in Pit-1 cells (Aipfx/fx:rGHRHRcre) increased pituitary tumor incidence and also accelerated tumor progression, supporting a loss-of-function/loss-of-heterozygosity model of tumorigenesis. Tumor development exhibited sexual dimorphism in wildtype and Aipfx/fx:rGHRHRcre animals. Despite the role of AHR as a tumor suppressor in other cancers, the observation that animals lacking AHR in all tissues, or ARNT in Pit-1 cells, do not develop somatotropinomas argues against the hypothesis that pituitary tumorigenesis in AIP-associated FIPA is related to decreased activities of either the Ahr or Arnt gene products.
Collapse
Affiliation(s)
- Anna L Shen
- The McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
| | - Susan M Moran
- The McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
| | - Edward N Glover
- The McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
| | - Bernice C Lin
- The McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
- Current address, Lin-Zhi International, 2945, Oakmead Village Court, Santa Clara, CA, 95051, United States
| | - Patrick R Carney
- The McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
| | - Christopher A Bradfield
- The McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
| |
Collapse
|
4
|
Kazzaz SA, Tawil J, Harhaj EW. The aryl hydrocarbon receptor-interacting protein in cancer and immunity: Beyond a chaperone protein for the dioxin receptor. J Biol Chem 2024; 300:107157. [PMID: 38479600 PMCID: PMC11002312 DOI: 10.1016/j.jbc.2024.107157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
The aryl hydrocarbon receptor (AhR)-interacting protein (AIP) is a ubiquitously expressed, immunophilin-like protein best known for its role as a co-chaperone in the AhR-AIP-Hsp90 cytoplasmic complex. In addition to regulating AhR and the xenobiotic response, AIP has been linked to various aspects of cancer and immunity that will be the focus of this review article. Loss-of-function AIP mutations are associated with pituitary adenomas, suggesting that AIP acts as a tumor suppressor in the pituitary gland. However, the tumor suppressor mechanisms of AIP remain unclear, and AIP can exert oncogenic functions in other tissues. While global deletion of AIP in mice yields embryonically lethal cardiac malformations, heterozygote, and tissue-specific conditional AIP knockout mice have revealed various physiological roles of AIP. Emerging studies have established the regulatory roles of AIP in both innate and adaptive immunity. AIP interacts with and inhibits the nuclear translocation of the transcription factor IRF7 to inhibit type I interferon production. AIP also interacts with the CARMA1-BCL10-MALT1 complex in T cells to enhance IKK/NF-κB signaling and T cell activation. Taken together, AIP has diverse functions that vary considerably depending on the client protein, the tissue, and the species.
Collapse
Affiliation(s)
- Sarah A Kazzaz
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA; Medical Scientist Training Program, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - John Tawil
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Edward W Harhaj
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
| |
Collapse
|
5
|
Rios EI, Hunsberger IL, Johnson JL. Insights into Hsp90 mechanism and in vivo functions learned from studies in the yeast, Saccharomyces cerevisiae. Front Mol Biosci 2024; 11:1325590. [PMID: 38389899 PMCID: PMC10881880 DOI: 10.3389/fmolb.2024.1325590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
The molecular chaperone Hsp90 (Heat shock protein, 90 kDa) is an abundant and essential cytosolic protein required for the stability and/or folding of hundreds of client proteins. Hsp90, along with helper cochaperone proteins, assists client protein folding in an ATP-dependent pathway. The laboratory of Susan Lindquist, in collaboration with other researchers, was the first to establish the yeast Saccharomyces cerevisiae as a model organism to study the functional interaction between Hsp90 and clients. Important insights from studies in her lab were that Hsp90 is essential, and that Hsp90 functions and cochaperone interactions are highly conserved between yeast and mammalian cells. Here, we describe key mechanistic insights into the Hsp90 folding cycle that were obtained using the yeast system. We highlight the early contributions of the laboratory of Susan Lindquist and extend our analysis into the broader use of the yeast system to analyze the understanding of the conformational cycle of Hsp90 and the impact of altered Hsp90 function on the proteome.
Collapse
Affiliation(s)
- Erick I Rios
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, ID, United States
| | - Isabel L Hunsberger
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, ID, United States
| | - Jill L Johnson
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, ID, United States
| |
Collapse
|
6
|
Roe SM, Török Z, McGown A, Horváth I, Spencer J, Pázmány T, Vigh L, Prodromou C. The Crystal Structure of the Hsp90-LA1011 Complex and the Mechanism by Which LA1011 May Improve the Prognosis of Alzheimer's Disease. Biomolecules 2023; 13:1051. [PMID: 37509087 PMCID: PMC10377191 DOI: 10.3390/biom13071051] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Functional changes in chaperone systems play a major role in the decline of cognition and contribute to neurological pathologies, such as Alzheimer's disease (AD). While such a decline may occur naturally with age or with stress or trauma, the mechanisms involved have remained elusive. The current models suggest that amyloid-β (Aβ) plaque formation leads to the hyperphosphorylation of tau by a Hsp90-dependent process that triggers tau neurofibrillary tangle formation and neurotoxicity. Several co-chaperones of Hsp90 can influence the phosphorylation of tau, including FKBP51, FKBP52 and PP5. In particular, elevated levels of FKBP51 occur with age and stress and are further elevated in AD. Recently, the dihydropyridine LA1011 was shown to reduce tau pathology and amyloid plaque formation in transgenic AD mice, probably through its interaction with Hsp90, although the precise mode of action is currently unknown. Here, we present a co-crystal structure of LA1011 in complex with a fragment of Hsp90. We show that LA1011 can disrupt the binding of FKBP51, which might help to rebalance the Hsp90-FKBP51 chaperone machinery and provide a favourable prognosis towards AD. However, without direct evidence, we cannot completely rule out effects on other Hsp90-co-chaprone complexes and the mechanisms they are involved in, including effects on Hsp90 client proteins. Nonetheless, it is highly significant that LA1011 showed promise in our previous AD mouse models, as AD is generally a disease affecting older patients, where slowing of disease progression could result in AD no longer being life limiting. The clinical value of LA1011 and its possible derivatives thereof remains to be seen.
Collapse
Affiliation(s)
- S Mark Roe
- Department of Biochemistry and Biomedicine, University of Sussex, Brighton BN1 9QG, UK
| | - Zsolt Török
- Institute of Biochemistry, Biological Research Centre, 6726 Szeged, Hungary
| | - Andrew McGown
- Sussex Drug Discovery Centre, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Ibolya Horváth
- Institute of Biochemistry, Biological Research Centre, 6726 Szeged, Hungary
| | - John Spencer
- Sussex Drug Discovery Centre, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Tamás Pázmány
- Gedeon Richter Plc, 1475 Budapest, Hungary
- National Vaccine Factory Plc, 4032 Debrecen, Hungary
| | - László Vigh
- Institute of Biochemistry, Biological Research Centre, 6726 Szeged, Hungary
| | | |
Collapse
|
7
|
Agrawal N, Gersey ZC, Abou-Al-Shaar H, Gardner PA, Mantica M, Agnihotri S, Mahmud H, Fazeli PK, Zenonos GA. Major Genetic Motifs in Pituitary Adenomas: A Practical Literature Update. World Neurosurg 2023; 169:43-50. [PMID: 36115566 PMCID: PMC11195535 DOI: 10.1016/j.wneu.2022.09.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND The literature includes many studies examining the genetic abnormalities that influence pituitary adenomas (PAs). We aimed to state the collective knowledge on the genetic underpinnings of PAs by organizing, summarizing, and consolidating the literature to serve as a comprehensive review for scientists and clinicians of the most up-to-date information underlying the genetic landscape of PAs. METHODS The PubMed and Google Scholar databases were searched using multiple key words and combined Medical Subject Headings terms; only articles published in English between January 2000 and January 2022 were included. Articles in which the focus did not relate to genetics, that included mainly anecdotal evidence, or that were single case studies were eliminated. RESULTS PAs are one of the most common intracranial neoplasms. However, the genetic underpinnings for these tumors are not yet fully elucidated. There are several categories of PAs: clinically significant versus not clinically significant, functional versus nonfunctional, and germline-derived versus sporadic origin. Each of these disease subcategories is characterized by unique genetic aberrations. Recently, more genes and other types of genetic aberrations have been identified as possible causes of PAs, such as copy number variations and altered levels of microRNAs. CONCLUSIONS This review serves to consolidate and summarize the literature discussing the genetic motifs of PAs to help physicians and scientists deliver patient-centered therapies.
Collapse
Affiliation(s)
- Nishant Agrawal
- Departments of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA; University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Zachary C Gersey
- Departments of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Hussam Abou-Al-Shaar
- Departments of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Paul A Gardner
- Departments of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Megan Mantica
- Departments of Neuro-Oncology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Sameer Agnihotri
- Departments of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Hussain Mahmud
- Departments of Endocrinology and Metabolism, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Pouneh K Fazeli
- Departments of Endocrinology and Metabolism, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Georgios A Zenonos
- Departments of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
8
|
Trofimiuk-Müldner M, Domagała B, Sokołowski G, Skalniak A, Hubalewska-Dydejczyk A. AIP gene germline variants in adult Polish patients with apparently sporadic pituitary macroadenomas. Front Endocrinol (Lausanne) 2023; 14:1098367. [PMID: 36843582 PMCID: PMC9950257 DOI: 10.3389/fendo.2023.1098367] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/23/2023] [Indexed: 02/12/2023] Open
Abstract
INTRODUCTION Up to 5% of all pituitary tumors are hereditary e.g. due to MEN1 or aryl hydrocarbon receptor-interacting protein (AIP) genes mutations. OBJECTIVES The study was aimed at the assessment of the frequency and characteristics of AIP-mutation related tumors in patients with apparently sporadic pituitary macroadenomas in the Polish population. MATERIALS AND METHODS The study included 131 patients (57 males, 74 females; median age 42 years) diagnosed with pituitary macroadenomas, and with a negative family history of familial isolated pituitary adenoma (FIPA) or multiple endocrine neoplasia type 1 (MEN1) syndromes. Sanger sequencing was used for the assessment of AIP gene variants. The study was approved by the Ethics Board of JUMC. RESULTS AIP variants were identified in five of the 131 included subjects (3.8%): one diagnosed with Cushing's disease, two with acromegaly, and two with non-secreting adenomas. Patients harboring hereditary AIP gene alterations did not differ from the rest of the study group in median age at diagnosis (41.0 vs. 42.5 years, P=0.8), median largest tumor diameter (25 vs. 24 mm, P=0.6), gender distribution (60.0% vs. 56.3% females, P=0.8), secreting tumor frequency (60.0% vs. 67.5%, P=0.7), or acromegaly diagnosis frequency (40.0% vs.37.3%, P=0.9). CONCLUSIONS In our series of apparently sporadic pituitary macroadenomas, AIP gene variant carriers did not differ substantially from patients with negative genetic testing. A risk factor-centred approach to AIP genetic screening may result in missing germline variants. Considering the clinical impact of such genetic variants and their relatively low penetrance, it is, however, doubtful if general genetic screening benefits the whole cohort of pituitary macroadenoma patients and their families.
Collapse
Affiliation(s)
- Małgorzata Trofimiuk-Müldner
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Kraków, Poland
- *Correspondence: Małgorzata Trofimiuk-Müldner,
| | - Bartosz Domagała
- Department of Endocrinology, Endocrine Oncology and Nuclear Medicine, University Hospital in Kraków, Kraków, Poland
| | - Grzegorz Sokołowski
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Kraków, Poland
| | - Anna Skalniak
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Kraków, Poland
| | | |
Collapse
|
9
|
Wang ZW, Niu L, Riaz S. Regulation of Ryanodine Receptor-Dependent Neurotransmitter Release by AIP, Calstabins, and Presenilins. ADVANCES IN NEUROBIOLOGY 2023; 33:287-304. [PMID: 37615871 DOI: 10.1007/978-3-031-34229-5_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Ryanodine receptors (RyRs) are Ca2+ release channels located in the endoplasmic reticulum membrane. Presynaptic RyRs play important roles in neurotransmitter release and synaptic plasticity. Recent studies suggest that the proper function of presynaptic RyRs relies on several regulatory proteins, including aryl hydrocarbon receptor-interacting protein, calstabins, and presenilins. Dysfunctions of these regulatory proteins can greatly impact neurotransmitter release and synaptic plasticity by altering the function or expression of RyRs. This chapter aims to describe the interaction between these proteins and RyRs, elucidating their crucial role in regulating synaptic function.
Collapse
Affiliation(s)
- Zhao-Wen Wang
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA.
| | - Longgang Niu
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Sadaf Riaz
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
10
|
Prodromou C, Aran-Guiu X, Oberoi J, Perna L, Chapple JP, van der Spuy J. HSP70-HSP90 Chaperone Networking in Protein-Misfolding Disease. Subcell Biochem 2023; 101:389-425. [PMID: 36520314 DOI: 10.1007/978-3-031-14740-1_13] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Molecular chaperones and their associated co-chaperones are essential in health and disease as they are key facilitators of protein-folding, quality control and function. In particular, the heat-shock protein (HSP) 70 and HSP90 molecular chaperone networks have been associated with neurodegenerative diseases caused by aberrant protein-folding. The pathogenesis of these disorders usually includes the formation of deposits of misfolded, aggregated protein. HSP70 and HSP90, plus their co-chaperones, have been recognised as potent modulators of misfolded protein toxicity, inclusion formation and cell survival in cellular and animal models of neurodegenerative disease. Moreover, these chaperone machines function not only in folding but also in proteasome-mediated degradation of neurodegenerative disease proteins. This chapter gives an overview of the HSP70 and HSP90 chaperones, and their respective regulatory co-chaperones, and explores how the HSP70 and HSP90 chaperone systems form a larger functional network and its relevance to counteracting neurodegenerative disease associated with misfolded proteins and disruption of proteostasis.
Collapse
Affiliation(s)
| | - Xavi Aran-Guiu
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, UK
| | - Jasmeen Oberoi
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, UK
| | - Laura Perna
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - J Paul Chapple
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | | |
Collapse
|
11
|
Gruszczyk J, Grandvuillemin L, Lai-Kee-Him J, Paloni M, Savva CG, Germain P, Grimaldi M, Boulahtouf A, Kwong HS, Bous J, Ancelin A, Bechara C, Barducci A, Balaguer P, Bourguet W. Cryo-EM structure of the agonist-bound Hsp90-XAP2-AHR cytosolic complex. Nat Commun 2022; 13:7010. [PMID: 36385050 PMCID: PMC9668932 DOI: 10.1038/s41467-022-34773-w] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/07/2022] [Indexed: 11/17/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor that mediates a broad spectrum of (patho)physiological processes in response to numerous substances including pollutants, natural products and metabolites. However, the scarcity of structural data precludes understanding of how AHR is activated by such diverse compounds. Our 2.85 Å structure of the human indirubin-bound AHR complex with the chaperone Hsp90 and the co-chaperone XAP2, reported herein, reveals a closed conformation Hsp90 dimer with AHR threaded through its lumen and XAP2 serving as a brace. Importantly, we disclose the long-awaited structure of the AHR PAS-B domain revealing a unique organisation of the ligand-binding pocket and the structural determinants of ligand-binding specificity and promiscuity of the receptor. By providing structural details of the molecular initiating event leading to AHR activation, our study rationalises almost forty years of biochemical data and provides a framework for future mechanistic studies and structure-guided drug design.
Collapse
Affiliation(s)
- Jakub Gruszczyk
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France.
| | - Loïc Grandvuillemin
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France
| | - Josephine Lai-Kee-Him
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France
| | - Matteo Paloni
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France
| | - Christos G Savva
- Leicester Institute of Structural & Chemical Biology and Department of Molecular & Cell Biology, University of Leicester, Leicester, UK
| | - Pierre Germain
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France
| | - Marina Grimaldi
- IRCM (Institut de Recherche en Cancérologie de Montpellier), Inserm U1194, Univ Montpellier, ICM, Montpellier, France
| | - Abdelhay Boulahtouf
- IRCM (Institut de Recherche en Cancérologie de Montpellier), Inserm U1194, Univ Montpellier, ICM, Montpellier, France
| | - Hok-Sau Kwong
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France
| | - Julien Bous
- Section of Receptor Biology & Signaling, Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Aurélie Ancelin
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France
| | - Cherine Bechara
- IGF, University of Montpellier, CNRS, Inserm, Montpellier, France
- Institut Universitaire de France (IUF), Paris, France
| | - Alessandro Barducci
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France
| | - Patrick Balaguer
- IRCM (Institut de Recherche en Cancérologie de Montpellier), Inserm U1194, Univ Montpellier, ICM, Montpellier, France
| | - William Bourguet
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France.
| |
Collapse
|
12
|
Advances towards Understanding the Mechanism of Action of the Hsp90 Complex. Biomolecules 2022; 12:biom12050600. [PMID: 35625528 PMCID: PMC9138868 DOI: 10.3390/biom12050600] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/13/2022] [Accepted: 04/17/2022] [Indexed: 12/24/2022] Open
Abstract
Hsp90 (Heat Shock Protein 90) is an ATP (Adenosine triphosphate) molecular chaperone responsible for the activation and maturation of client proteins. The mechanism by which Hsp90 achieves such activation, involving structurally diverse client proteins, has remained enigmatic. However, recent advances using structural techniques, together with advances in biochemical studies, have not only defined the chaperone cycle but have shed light on its mechanism of action. Hsp90 hydrolysis of ATP by each protomer may not be simultaneous and may be dependent on the specific client protein and co-chaperone complex involved. Surprisingly, Hsp90 appears to remodel client proteins, acting as a means by which the structure of the client protein is modified to allow its subsequent refolding to an active state, in the case of kinases, or by making the client protein competent for hormone binding, as in the case of the GR (glucocorticoid receptor). This review looks at selected examples of client proteins, such as CDK4 (cyclin-dependent kinase 4) and GR, which are activated according to the so-called ‘remodelling hypothesis’ for their activation. A detailed description of these activation mechanisms is paramount to understanding how Hsp90-associated diseases develop.
Collapse
|
13
|
Johnson JL. Mutations in Hsp90 Cochaperones Result in a Wide Variety of Human Disorders. Front Mol Biosci 2021; 8:787260. [PMID: 34957217 PMCID: PMC8694271 DOI: 10.3389/fmolb.2021.787260] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/08/2021] [Indexed: 12/19/2022] Open
Abstract
The Hsp90 molecular chaperone, along with a set of approximately 50 cochaperones, mediates the folding and activation of hundreds of cellular proteins in an ATP-dependent cycle. Cochaperones differ in how they interact with Hsp90 and their ability to modulate ATPase activity of Hsp90. Cochaperones often compete for the same binding site on Hsp90, and changes in levels of cochaperone expression that occur during neurodegeneration, cancer, or aging may result in altered Hsp90-cochaperone complexes and client activity. This review summarizes information about loss-of-function mutations of individual cochaperones and discusses the overall association of cochaperone alterations with a broad range of diseases. Cochaperone mutations result in ciliary or muscle defects, neurological development or degeneration disorders, and other disorders. In many cases, diseases were linked to defects in established cochaperone-client interactions. A better understanding of the functional consequences of defective cochaperones will provide new insights into their functions and may lead to specialized approaches to modulate Hsp90 functions and treat some of these human disorders.
Collapse
Affiliation(s)
- Jill L Johnson
- Department of Biological Sciences and Center for Reproductive Biology, University of Idaho, Moscow, ID, United States
| |
Collapse
|
14
|
Garcia-Rendueles AR, Chenlo M, Oroz-Gonjar F, Solomou A, Mistry A, Barry S, Gaston-Massuet C, Garcia-Lavandeira M, Perez-Romero S, Suarez-Fariña M, Pradilla-Dieste A, Dieguez C, Mehlen P, Korbonits M, Alvarez CV. RET signalling provides tumorigenic mechanism and tissue specificity for AIP-related somatotrophinomas. Oncogene 2021; 40:6354-6368. [PMID: 34588620 PMCID: PMC8585666 DOI: 10.1038/s41388-021-02009-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 08/21/2021] [Accepted: 09/06/2021] [Indexed: 02/07/2023]
Abstract
It is unclear how loss-of-function germline mutations in the widely-expressed co-chaperone AIP, result in young-onset growth hormone secreting pituitary tumours. The RET receptor, uniquely co-expressed in somatotrophs with PIT1, induces apoptosis when unliganded, while RET supports cell survival when it is bound to its ligand. We demonstrate that at the plasma membrane, AIP is required to form a complex with monomeric-intracellular-RET, caspase-3 and PKCδ resulting in PIT1/CDKN2A-ARF/p53-apoptosis pathway activation. AIP-deficiency blocks RET/caspase-3/PKCδ activation preventing PIT1 accumulation and apoptosis. The presence or lack of the inhibitory effect on RET-induced apoptosis separated pathogenic AIP variants from non-pathogenic ones. We used virogenomics in neonatal rats to demonstrate the effect of mutant AIP protein on the RET apoptotic pathway in vivo. In adult male rats altered AIP induces elevated IGF-1 and gigantism, with pituitary hyperplasia through blocking the RET-apoptotic pathway. In females, pituitary hyperplasia is induced but IGF-1 rise and gigantism are blunted by puberty. Somatotroph adenomas from pituitary-specific Aip-knockout mice overexpress the RET-ligand GDNF, therefore, upregulating the survival pathway. Somatotroph adenomas from patients with or without AIP mutation abundantly express GDNF, but AIP-mutated tissues have less CDKN2A-ARF expression. Our findings explain the tissue-specific mechanism of AIP-induced somatotrophinomas and provide a previously unknown tumorigenic mechanism, opening treatment avenues for AIP-related tumours.
Collapse
Affiliation(s)
- Angela R Garcia-Rendueles
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Miguel Chenlo
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Fernando Oroz-Gonjar
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Antonia Solomou
- Department of Endocrinology, William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Anisha Mistry
- Department of Endocrinology, William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sayka Barry
- Department of Endocrinology, William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Carles Gaston-Massuet
- Department of Endocrinology, William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Montserrat Garcia-Lavandeira
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Sihara Perez-Romero
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Maria Suarez-Fariña
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Alberto Pradilla-Dieste
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Carlos Dieguez
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Patrick Mehlen
- Patrick Mehlen, Apoptosis, Cancer and Development Laboratory- Equipe labellisée 'La Ligue', LabEx DEVweCAN, Institut PLAsCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, 69008, Lyon, France
| | - Márta Korbonits
- Department of Endocrinology, William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Clara V Alvarez
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain.
| |
Collapse
|
15
|
Dean ME, Johnson JL. Human Hsp90 cochaperones: perspectives on tissue-specific expression and identification of cochaperones with similar in vivo functions. Cell Stress Chaperones 2021; 26:3-13. [PMID: 33037995 PMCID: PMC7736379 DOI: 10.1007/s12192-020-01167-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/12/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
The Hsp90 molecular chaperone is required for the function of hundreds of different cellular proteins. Hsp90 and a cohort of interacting proteins called cochaperones interact with clients in an ATP-dependent cycle. Cochaperone functions include targeting clients to Hsp90, regulating Hsp90 ATPase activity, and/or promoting Hsp90 conformational changes as it progresses through the cycle. Over the last 20 years, the list of cochaperones identified in human cells has grown from the initial six identified in complex with steroid hormone receptors and protein kinases to about fifty different cochaperones found in Hsp90-client complexes. These cochaperones may be placed into three groups based on shared Hsp90 interaction domains. Available evidence indicates that cochaperones vary in client specificity, abundance, and tissue distribution. Many of the cochaperones have critical roles in regulation of cancer and neurodegeneration. A more limited set of cochaperones have cellular functions that may be limited to tissues such as muscle and testis. It is likely that a small set of cochaperones are part of the core Hsp90 machinery required for the folding of a wide range of clients. The presence of more selective cochaperones may allow greater control of Hsp90 activities across different tissues or during development.
Collapse
Affiliation(s)
- Marissa E Dean
- Department of Biological Sciences, University of Idaho, Moscow, ID, 83844-3051, USA
| | - Jill L Johnson
- Department of Biological Sciences, University of Idaho, Moscow, ID, 83844-3051, USA.
- Center for Reproductive Biology, University of Idaho, Moscow, ID, 83844-3051, USA.
| |
Collapse
|
16
|
Dal J, Nielsen EH, Klose M, Feldt-Rasmussen U, Andersen M, Vang S, Korbonits M, Jørgensen JOL. Phenotypic and genotypic features of a large kindred with a germline AIP variant. Clin Endocrinol (Oxf) 2020; 93:146-153. [PMID: 32324286 DOI: 10.1111/cen.14207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 11/30/2022]
Abstract
CONTEXT Acromegaly is usually a sporadic disease, but familial cases occur. Mutations in the aryl hydrocarbon receptor-interacting protein (AIP) gene are associated with familial pituitary adenoma predisposition. However, the pathogenicity of some AIP variants remains unclear and additional unknown genes may be involved. OBJECTIVE To explore the phenotype and genotype of a large kindred carrying the p.R304Q AIP variant. METHODS The family comprised 52 family members at risk of carrying the p.R304Q AIP variant including a case with gigantism and one with acromegaly and several family members with acromegalic features. Nine family members (three trios) underwent exome sequencing to identify putative pathogenic variants. RESULTS We identified 31 p.R304Q carriers, and based on two cases with somatotropinomas, the disease penetrance was 6%. We observed physical signs of acromegaly in several family members, which were independent of AIP status. Serum insulin-like growth factor-I (IGF-I) levels in all family members were above the mean for age and sex (IGF-I SDS: +0.6 [CI95% +0.4-0.9], P < .01). Exome analysis identified two candidate genes: PDE11A, known to be associated with the development of adrenal tumours, and ALG14. Ten asymptomatic p.R304Q family members (age >50 years) were screened for the PDE11A and ALG14 variant; both variants were present in five of ten persons. CONCLUSIONS This large family adds new information on the p.R304Q AIP variant, and data suggest two new candidate genes could be associated with growth hormone excess.
Collapse
Affiliation(s)
- Jakob Dal
- Department of Endocrinology, Aalborg University Hospital, Aalborg, Denmark
- Steno Diabetic Center Northjutland, Aalborg, Denmark
| | - Eigil H Nielsen
- Department of Endocrinology, Aalborg University Hospital, Aalborg, Denmark
| | - Marianne Klose
- Department of Endocrinology and Metabolism, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ulla Feldt-Rasmussen
- Department of Endocrinology and Metabolism, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marianne Andersen
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Søren Vang
- Department of Molecular Medicine, Aarhus University Hospital, Denmark
| | - Márta Korbonits
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Jens Otto L Jørgensen
- Department of Endocrinology and Internal Medicine and Medical Research Laboratories, Aarhus University Hospital, Aarhus C, Denmark
| |
Collapse
|
17
|
Vitali E, Boemi I, Piccini S, Tarantola G, Smiroldo V, Lavezzi E, Brambilla T, Zerbi A, Carnaghi C, Mantovani G, Spada A, Lania AG. A novel insight into the anticancer mechanism of metformin in pancreatic neuroendocrine tumor cells. Mol Cell Endocrinol 2020; 509:110803. [PMID: 32251713 DOI: 10.1016/j.mce.2020.110803] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 03/24/2020] [Accepted: 03/29/2020] [Indexed: 12/12/2022]
Abstract
The antidiabetic drug metformin displays anticancer properties in several neoplasms. In pituitary NETs, aryl hydrocarbon receptor-interacting protein (AIP) is up-regulated by the somatostatin analog octreotide. Metformin inhibited QGP-1 cell proliferation in a dose- and time-dependent manner, at concentrations similar to those achievable in treated patients (-31 ± 12%, p < 0.05 vs basal at 100 μM). Moreover, metformin decreased pancreatic neuroendocrine tumors (PAN-NETs) cell proliferation (-62 ± 15%, p < 0.0001 vs basal at 10 mM), without any additive effect when combined with octreotide. Both octreotide and metformin induced AIP up-regulation. AIP silencing abolished the reduction of mTOR phosphorylation induced by metformin and octreotide. Moreover, metformin decreased HSP70, increased Zac1 and AhR expression; these effects were abolished in AIP silenced QGP-1 cells. In conclusion, metformin acts as an anticancer agent in PAN-NET cells, its activity is mediated by AIP and its interacting proteins. These findings provide a novel insight into the antitumorigenic mechanism of metformin.
Collapse
Affiliation(s)
- E Vitali
- Laboratory of Cellular and Molecular Endocrinology, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.
| | - I Boemi
- Laboratory of Cellular and Molecular Endocrinology, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - S Piccini
- Endocrinology and Diabetology Unit Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - G Tarantola
- Laboratory of Cellular and Molecular Endocrinology, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - V Smiroldo
- Oncology Unit, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - E Lavezzi
- Endocrinology and Diabetology Unit Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - T Brambilla
- Department of Pathology, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - A Zerbi
- Pancreas Surgery Unit, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - C Carnaghi
- Oncology Unit, Bolzano Hospital, Bolzano, Italy
| | - G Mantovani
- Endocrinology and Diabetology Unit, IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - A Spada
- Endocrinology and Diabetology Unit, IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - A G Lania
- Laboratory of Cellular and Molecular Endocrinology, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy; Endocrinology and Diabetology Unit Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| |
Collapse
|
18
|
Schernthaner-Reiter MH, Trivellin G, Stratakis CA. Chaperones, somatotroph tumors and the cyclic AMP (cAMP)-dependent protein kinase (PKA) pathway. Mol Cell Endocrinol 2020; 499:110607. [PMID: 31586652 DOI: 10.1016/j.mce.2019.110607] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 02/08/2023]
Abstract
The cAMP-PKA pathway plays an essential role in the pituitary gland, governing cell differentiation and survival, and maintenance of endocrine function. Somatotroph growth hormone transcription and release as well as cell proliferation are regulated by the cAMP-PKA pathway; cAMP-PKA pathway abnormalities are frequently detected in sporadic as well as in hereditary somatotroph tumors and more rarely in other pituitary tumors. Inactivating variants of the aryl hydrocarbon receptor-interacting protein (AIP)-coding gene are the genetic cause of a subset of familial isolated pituitary adenomas (FIPA). Multiple functional links between the co-chaperone AIP and the cAMP-PKA pathway have been described. This review explores the role of chaperones including AIP in normal pituitary function as well as in somatotroph tumors, and their interaction with the cAMP-PKA pathway.
Collapse
Affiliation(s)
| | - Giampaolo Trivellin
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, USA
| | - Constantine A Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, USA
| |
Collapse
|
19
|
Re A, Ferraù F, Cafiero C, Spagnolo F, Barresi V, Romeo DP, Ragonese M, Grassi C, Pontecorvi A, Farsetti A, Cannavò S. Somatic Deletion in Exon 10 of Aryl Hydrocarbon Receptor Gene in Human GH-Secreting Pituitary Tumors. Front Endocrinol (Lausanne) 2020; 11:591039. [PMID: 33281746 PMCID: PMC7689685 DOI: 10.3389/fendo.2020.591039] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE/PURPOSE The aryl hydrocarbon receptor (AHR) pathway plays a critical role in the biology of Growth Hormone (GH)-secreting pituitary tumor (somatotropinoma). Germline rs2066853 AHR variant was found to be more frequent among acromegaly patients and associated with a more severe disease with larger invasive somatropinoma, and with resistance to somatostatin analogs treatment in patients living in polluted areas. However, no somatic changes in AHR gene have been reported so far in acromegaly patients. On that basis, the aim of the study was to assess at the somatic level the AHR gene status encompassing exon 10 region, also because of the high rate of variants found in this genomic region. METHODS A cohort of 13 patients aged 20-76 years with biochemical, clinical and histological diagnosis of somatotropinoma was studied. DNA and RNA from pituitary tumor histological samples have been extracted and analyzed by PCR and direct sequencing for AHR gene variants, and compared with corresponding patients' germline DNA as well as normal pituitary tissue as reference control. RESULTS A degenerated letter codes in the region corresponding to AHR exon 10 (c.1239-c.2056) was detected in somatotropinomas-derived DNA but not in that of matched germline and pituitary normal tissue. By multiple PCR and sequencing analysis, we observed amplification only before codon 1246 and after codon 1254, confirming the presence of a tumor-restricted somatic deletion in the 5' upstream region of AHR exon 10. Analysis of PCR-amplified cDNA revealed a wildtype sequence of exon 9 and 10 in normal pituitary tissue, and a wildtype sequence of exon 9 and 10 up to codon 1246 and no sequence after the deletion region (c.1246-c.1254) in 6 out of 9 tumor samples. Patients carrying the germline rs2066853 AHR variant showed no somatic LOH at the corresponding genetic locus. CONCLUSION This is the first demonstration of a recurrent somatic deletion in the exon 10 of the AHR gene in somatotropinomas. The functional impact of this genetic finding needs to be clarified.
Collapse
Affiliation(s)
- Agnese Re
- Institute for Systems Analysis and Computer Science “A. Ruberti” (IASI), National Research Council (CNR), Rome, Italy
- *Correspondence: Salvatore Cannavò, ; Agnese Re,
| | - Francesco Ferraù
- Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
- Endocrine Unit, University Hospital G. Martino, Messina, Italy
| | | | | | - Valeria Barresi
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | | | - Marta Ragonese
- Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
| | - Claudio Grassi
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alfredo Pontecorvi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonella Farsetti
- Institute for Systems Analysis and Computer Science “A. Ruberti” (IASI), National Research Council (CNR), Rome, Italy
| | - Salvatore Cannavò
- Department of Human Pathology of Adulthood and Childhood, University of Messina, Messina, Italy
- Endocrine Unit, University Hospital G. Martino, Messina, Italy
- *Correspondence: Salvatore Cannavò, ; Agnese Re,
| |
Collapse
|
20
|
Schernthaner-Reiter MH, Trivellin G, Stratakis CA. Interaction of AIP with protein kinase A (cAMP-dependent protein kinase). Hum Mol Genet 2019; 27:2604-2613. [PMID: 29726992 DOI: 10.1093/hmg/ddy166] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/29/2018] [Accepted: 05/01/2018] [Indexed: 01/07/2023] Open
Abstract
Germline mutations in the aryl hydrocarbon receptor-interacting protein (AIP) gene cause mostly somatotropinomas and/or prolactinomas in a subset of familial isolated pituitary adenomas (FIPA). AIP has been shown to interact with phosphodiesterases (PDEs) and G proteins, suggesting a link to the cyclic AMP (cAMP)-dependent protein kinase (PKA) pathway. Upregulation of PKA is seen in sporadic somatotropinomas that carry GNAS mutations, and those in Carney complex that are due to PRKAR1A mutations. To elucidate the mechanism of AIP-dependent pituitary tumorigenesis, we studied potential functional and physical interactions of AIP with PKA's main subunits PRKAR1A (R1α) and PRKACA (Cα). We found that AIP physically interacts with both R1α and Cα; this interaction is enhanced when all three components are present, but maintained during Cα-R1α dissociation by PKA activation, indicating that AIP binds Cα/R1α both in complex and separately. The interaction between AIP and R1α/Cα is reduced when the frequent AIP pathogenic mutation p.R304* is present. AIP protein levels are regulated both by translation and the ubiquitin/proteasome pathway and Cα stabilizes both AIP and R1α protein levels. AIP reduction by siRNA leads to an increase of PKA activity, which is disproportionately enhanced during PDE4-inhibition. We show that AIP interacts with the PKA pathway on multiple levels, including a physical interaction with both the main regulatory (R1α) and catalytic (Cα) PKA subunits and a functional interaction with PDE4-dependent PKA activation. These findings provide novel insights on the mechanisms of AIP-dependent pituitary tumorigenesis.
Collapse
Affiliation(s)
- Marie Helene Schernthaner-Reiter
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Giampaolo Trivellin
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Constantine A Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
21
|
Yadav RP, Boyd K, Yu L, Artemyev NO. Interaction of the tetratricopeptide repeat domain of aryl hydrocarbon receptor-interacting protein-like 1 with the regulatory Pγ subunit of phosphodiesterase 6. J Biol Chem 2019; 294:15795-15807. [PMID: 31488544 DOI: 10.1074/jbc.ra119.010666] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/03/2019] [Indexed: 12/13/2022] Open
Abstract
Phosphodiesterase-6 (PDE6) is key to both phototransduction and health of rods and cones. Proper folding of PDE6 relies on the chaperone activity of aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1), and mutations in both PDE6 and AIPL1 can cause a severe form of blindness. Although AIPL1 and PDE6 are known to interact via the FK506-binding protein domain of AIPL1, the contribution of the tetratricopeptide repeat (TPR) domain of AIPL1 to its chaperone function is poorly understood. Here, we demonstrate that AIPL1-TPR interacts specifically with the regulatory Pγ subunit of PDE6. Use of NMR chemical shift perturbation (CSP) mapping technique revealed the interface between the C-terminal portion of Pγ and AIPL1-TPR. Our solution of the crystal structure of the AIPL1-TPR domain provided additional information, which together with the CSP data enabled us to generate a model of this interface. Biochemical analysis of chimeric AIPL1-AIP proteins supported this model and also revealed a correlation between the affinity of AIPL1-TPR for Pγ and the ability of Pγ to potentiate the chaperone activity of AIPL1. Based on these results, we present a model of the larger AIPL1-PDE6 complex. This supports the importance of simultaneous interactions of AIPL1-FK506-binding protein with the prenyl moieties of PDE6 and AIPL1-TPR with the Pγ subunit during the folding and/or assembly of PDE6. This study sheds new light on the versatility of TPR domains in protein folding by describing a novel TPR-protein binding partner, Pγ, and revealing that this subunit imparts AIPL1 selectivity for its client.
Collapse
Affiliation(s)
- Ravi P Yadav
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Kimberly Boyd
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Liping Yu
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242.,NMR Core Facility, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Nikolai O Artemyev
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 .,Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| |
Collapse
|
22
|
Stojanovic M, Wu Z, Stiles CE, Miljic D, Soldatovic I, Pekic S, Doknic M, Petakov M, Popovic V, Strasburger C, Korbonits M. Circulating aryl hydrocarbon receptor-interacting protein (AIP) is independent of GH secretion. Endocr Connect 2019; 8:326-337. [PMID: 30830858 PMCID: PMC6432870 DOI: 10.1530/ec-19-0082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/04/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Aryl hydrocarbon receptor-interacting protein (AIP) is evolutionarily conserved and expressed widely throughout the organism. Loss-of-function AIP mutations predispose to young-onset pituitary adenomas. AIP co-localizes with growth hormone in normal and tumorous somatotroph secretory vesicles. AIP protein is detectable in circulation. We aimed to investigate possible AIP and GH co-secretion, by studying serum AIP and GH levels at baseline and after GH stimulation or suppression, in GH deficiency (GHD) and in acromegaly patients. SUBJECTS AND METHODS Insulin tolerance test (ITT) was performed in GHD patients (n = 13) and age-BMI-matched normal GH axis control patients (n = 31). Oral glucose tolerance test (OGTT) was performed in active acromegaly patients (n = 26) and age-BMI-matched normal GH axis control patients (n = 18). In-house immunometric assay was developed for measuring circulating AIP. RESULTS Serum AIP levels were in the 0.1 ng/mL range independently of gender, age or BMI. Baseline AIP did not differ between GHD and non-GHD or between acromegaly and patients with no acromegaly. There was no change in peak, trough or area under the curve during OGTT or ITT. Serum AIP did not correlate with GH during ITT or OGTT. CONCLUSIONS Human circulating serum AIP in vivo was assessed by a novel immunometric assay. AIP levels were independent of age, sex or BMI and unaffected by hypoglycaemia or hyperglycaemia. Despite co-localization in secretory vesicles, AIP and GH did not correlate at baseline or during GH stimulation or suppression tests. A platform of reliable serum AIP measurement is established for further research of its circulatory source, role and impact.
Collapse
Affiliation(s)
- Marko Stojanovic
- Neuroendocrinology Department, Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Centre of Serbia, Belgrade, Serbia
- University of Belgrade, Medical Faculty, Belgrade, Serbia
| | - Zida Wu
- Department of Medicine for Endocrinology, Diabetes and Nutritional Medicine, Charité Universitätsmedizin, Campus Mitte, Berlin, Germany
| | - Craig E Stiles
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Dragana Miljic
- Neuroendocrinology Department, Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Centre of Serbia, Belgrade, Serbia
- University of Belgrade, Medical Faculty, Belgrade, Serbia
| | - Ivan Soldatovic
- University of Belgrade, Medical Faculty, Belgrade, Serbia
- Insitute of Medical Statistics and Informatics, Belgrade, Serbia
| | - Sandra Pekic
- Neuroendocrinology Department, Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Centre of Serbia, Belgrade, Serbia
- University of Belgrade, Medical Faculty, Belgrade, Serbia
| | - Mirjana Doknic
- Neuroendocrinology Department, Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Centre of Serbia, Belgrade, Serbia
- University of Belgrade, Medical Faculty, Belgrade, Serbia
| | - Milan Petakov
- Neuroendocrinology Department, Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Centre of Serbia, Belgrade, Serbia
- University of Belgrade, Medical Faculty, Belgrade, Serbia
| | - Vera Popovic
- University of Belgrade, Medical Faculty, Belgrade, Serbia
| | - Christian Strasburger
- Department of Medicine for Endocrinology, Diabetes and Nutritional Medicine, Charité Universitätsmedizin, Campus Mitte, Berlin, Germany
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
23
|
Yu L, Yadav RP, Artemyev NO. NMR resonance assignments of the TPR domain of human aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1). BIOMOLECULAR NMR ASSIGNMENTS 2019; 13:79-83. [PMID: 30341566 PMCID: PMC6440825 DOI: 10.1007/s12104-018-9856-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/14/2018] [Indexed: 06/08/2023]
Abstract
Aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) is a photoreceptor-specific chaperone of phosphodiesterase-6, a key effector enzyme in the phototransduction cascade. It contains an N-terminal FK506-binding protein (FKBP) domain and a C-terminal tetratricopeptide repeat (TPR) domain. Mutations in AIPL1, including many missense mutations in both FKBP and TPR domains, have been associated with Leber congenital amaurosis, a severe inherited retinopathy that causes blindness. TPR-domain containing proteins are known to interact with HSP90. However, the structure of AIPL1-TPR domain is presently not determined and little is known about the contribution of the TPR domain to the chaperone function of AIPL1. Here, we report the backbone and sidechain assignments of the TPR domain of AIPL1. These assignments reveal that AIPL1-TPR is an α-helical protein containing seven α-helices connected via short loops. Peak broadening or structural disorder is observed for a cluster of hydrophobic residues of W218, W222 and L223. Therefore, these assignments provide a framework for further structural determination of AIPL1-TPR domain and its interactions with various binding partners for elucidation of the mechanism of TPR contribution to the chaperone function of AIPL1.
Collapse
Affiliation(s)
- Liping Yu
- Department of Biochemistry, University of Iowa Carver College of Medicine, B291 CBRB, 285 Newton Road, Iowa City, IA, 52242, USA.
- CCOM NMR Core Facility, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| | - Ravi P Yadav
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, 5-532 BSB, 51 Newton Road, Iowa City, IA, 52242, USA
| | - Nikolai O Artemyev
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, 5-532 BSB, 51 Newton Road, Iowa City, IA, 52242, USA.
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| |
Collapse
|
24
|
Pepe S, Korbonits M, Iacovazzo D. Germline and mosaic mutations causing pituitary tumours: genetic and molecular aspects. J Endocrinol 2019; 240:R21-R45. [PMID: 30530903 DOI: 10.1530/joe-18-0446] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 11/07/2018] [Indexed: 12/24/2022]
Abstract
While 95% of pituitary adenomas arise sporadically without a known inheritable predisposing mutation, in about 5% of the cases they can arise in a familial setting, either isolated (familial isolated pituitary adenoma or FIPA) or as part of a syndrome. FIPA is caused, in 15-30% of all kindreds, by inactivating mutations in the AIP gene, encoding a co-chaperone with a vast array of interacting partners and causing most commonly growth hormone excess. While the mechanisms linking AIP with pituitary tumorigenesis have not been fully understood, they are likely to involve several pathways, including the cAMP-dependent protein kinase A pathway via defective G inhibitory protein signalling or altered interaction with phosphodiesterases. The cAMP pathway is also affected by other conditions predisposing to pituitary tumours, including X-linked acrogigantism caused by duplications of the GPR101 gene, encoding an orphan G stimulatory protein-coupled receptor. Activating mosaic mutations in the GNAS gene, coding for the Gα stimulatory protein, cause McCune-Albright syndrome, while inactivating mutations in the regulatory type 1α subunit of protein kinase A represent the most frequent genetic cause of Carney complex, a syndromic condition with multi-organ manifestations also involving the pituitary gland. In this review, we discuss the genetic and molecular aspects of isolated and syndromic familial pituitary adenomas due to germline or mosaic mutations, including those secondary to AIP and GPR101 mutations, multiple endocrine neoplasia type 1 and 4, Carney complex, McCune-Albright syndrome, DICER1 syndrome and mutations in the SDHx genes underlying the association of familial paragangliomas and phaeochromocytomas with pituitary adenomas.
Collapse
Affiliation(s)
- Sara Pepe
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| | - Donato Iacovazzo
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| |
Collapse
|
25
|
Bizzi MF, Bolger GB, Korbonits M, Ribeiro-Oliveira Jr. A. Phosphodiesterases and cAMP Pathway in Pituitary Diseases. Front Endocrinol (Lausanne) 2019; 10:141. [PMID: 30941100 PMCID: PMC6433792 DOI: 10.3389/fendo.2019.00141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/15/2019] [Indexed: 12/21/2022] Open
Abstract
Human phosphodiesterases (PDEs) comprise a complex superfamily of enzymes derived from 24 genes separated into 11 PDE gene families (PDEs 1-11), expressed in different tissues and cells, including heart and brain. The isoforms PDE4, PDE7, and PDE8 are specific for the second messenger cAMP, which is responsible for mediating diverse physiological actions involving different hormones and neurotransmitters. The cAMP pathway plays an important role in the development and function of endocrine tissues while phosphodiesterases are responsible for ensuring the appropriate intensity of the actions of this pathway by hydrolyzing cAMP to its inactive form 5'-AMP. PDE1, PDE2, PDE4, and PDE11A are highly expressed in the pituitary, and overexpression of some PDE4 isoforms have been demonstrated in different pituitary adenoma subtypes. This observed over-expression in pituitary adenomas, although of unknown etiology, has been considered a compensatory response to tumorigenesis. PDE4A4/5 has a unique interaction with the co-chaperone aryl hydrocarbon receptor-interacting protein (AIP), a protein implicated in somatotroph tumorigenesis via germline loss-of-function mutations. Based on the association of low PDE4A4 expression with germline AIP-mutation-positive samples, the available data suggest that lack of AIP hinders the upregulation of PDE4A4 protein seen in sporadic somatotrophinomas. This unique disturbance of the cAMP-PDE pathway observed in the majority of AIP-mutation positive adenomas could contribute to their well-described poor response to somatostatin analogs and may support a role in tumorigenesis.
Collapse
Affiliation(s)
- Mariana Ferreira Bizzi
- Department of Internal Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Graeme B. Bolger
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Márta Korbonits
- Center for Endocrinology, Barts and The London School of Medicine, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Antonio Ribeiro-Oliveira Jr.
- Department of Internal Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Antonio Ribeiro-Oliveira Jr.
| |
Collapse
|
26
|
Tuncer FN, Çiftçi Doğanşen S, Serbest E, Tanrıkulu S, Ekici Y, Bilgiç B, Yarman S. Screening of AIP Gene Variations in a Cohort of Turkish Patients with Young-Onset Sporadic Hormone-Secreting Pituitary Adenomas. Genet Test Mol Biomarkers 2018; 22:702-708. [PMID: 30461320 DOI: 10.1089/gtmb.2018.0133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aims: Aryl hydrocarbon receptor-interacting protein (AIP) gene mutations have long been associated with apparently sporadic pituitary adenomas (PAs) with a prevalence range of 0-12%. The aim of this study was to evaluate the frequency of germline AIP variations in a large cohort of apparently sporadic PAs diagnosed before the age of 40 years, who did not exhibit hypercalcemia and/or MEN1 syndrome components during long-term follow-up. Materials and Methods: A total of 97 patients, diagnosed with functional PAs ≤40 years old, composed of somatotropinoma (n = 55), prolactinoma (n = 25), and corticotrophinoma (n = 17), were recruited for this study. Fifty-one of these patients [somatotropinoma (n = 30), prolactinoma (n = 15), and corticotrophinoma (n = 11)] were previously reported as AIP mutation-negative by Sanger sequencing. The entire coding sequence of the AIP gene, along with exon/intron boundaries and the untranslated regions of 41 newly recruited patients, were sequenced for germline variations. In addition, all patients were subjected to multiplex ligation-dependent probe amplification to detect copy number variations in the AIP gene. Results: The AIP c.911G>A: p.Arg304Gln (rs104894190) variant was detected in only two patients with functional PA: one with somatotropinoma [in 1/55 (1.8%)] and one with prolactinoma [in 1/25 (4%)]. None of the corticotrophinomas revealed AIP gene alterations. Thus, the overall prevalence of AIP variation was 2.1% in our cohort. Conclusions: Germline AIP gene variations among Turkish patients with apparently sporadic PAs are relatively rare among patients ≤40 years old. None of the patients in our cohort revealed any obviously pathogenic AIP variants.
Collapse
Affiliation(s)
- Feyza Nur Tuncer
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Sema Çiftçi Doğanşen
- Division of Endocrinology and Metabolic Diseases, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.,Division of Endocrinology and Metabolic Diseases, Bakırköy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Esin Serbest
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Seher Tanrıkulu
- Division of Endocrinology and Metabolic Diseases, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.,Division of Endocrinology and Metabolic Diseases, Haydarpaşa Numune Training and Research Hospital, Istanbul, Turkey
| | - Yeliz Ekici
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Bilge Bilgiç
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sema Yarman
- Division of Endocrinology and Metabolic Diseases, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
27
|
Cai F, Hong Y, Xu J, Wu Q, Reis C, Yan W, Wang W, Zhang J. A Novel Mutation of Aryl Hydrocarbon Receptor Interacting Protein Gene Associated with Familial Isolated Pituitary Adenoma Mediates Tumor Invasion and Growth Hormone Hypersecretion. World Neurosurg 2018; 123:e45-e59. [PMID: 30447469 DOI: 10.1016/j.wneu.2018.11.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 11/02/2018] [Accepted: 11/04/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Germline mutations in the aryl hydrocarbon receptor-interacting protein (AIP) gene were identified in nearly 20% of families with familial isolated pituitary adenoma. Some variants of AIP have been confirmed to induce tumor cell proliferation and invasiveness; however, the mechanism is still unclear. METHODS A novel missense mutation (c.512C>T, p.T171I) was discovered in 3 patients from a Chinese family with familial isolated pituitary adenoma. In silico and multiplex ligation-dependent probe amplification analysis predicted the mutation to be pathogenic. GH3 and 293FT cell lines were used to verify the variant's effect on cell proliferation (Cell Counting Kit-8), invasiveness (Transwell) and growth hormone (GH) secretion (enzyme-linked immunosorbent assay) by transfection with different vectors: control, blank vector, wild-type AIP, p.T171I variant (experimental group), p.Q315* variant, and AIP small interfering RNA. Furthermore, Zac1, Sstr2, interleukin (IL)-6, and Stat3/phosphorylation-Stat3 expression (reverse transcription polymerase chain reaction, Western blot) in each group was also evaluated. RESULTS The experimental group, p.Q315* variant group, and AIP small interfering RNA-overexpressing group promoted cell proliferation at 24 and 48 hours, respectively (compared with the control group; P < 0.01 for both). Similarly, the cells in the experimental group manifested more invasion and GH secretion compared with the control group (P < 0.01 and P < 0.05, respectively). Furthermore, the experimental group cells expressed less Sstr2 (a prerequisite for the responsiveness to somatostatin analogues) and Zac1 (tumor suppressor gene), but more IL-6 and phosphorylated-Stat3 (GH-secretion related). CONCLUSIONS The novel AIP mutation c.512C>T (p.T171I) is a pathogenic variant that promoted cell proliferation, invasiveness, and GH secretion through regulation of Sstr2, Zac1, and IL-6/phosphorylated-Stat3 expression.
Collapse
Affiliation(s)
- Feng Cai
- Department of Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan Hong
- Department of Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jinghong Xu
- Department of Pathology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qun Wu
- Department of Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Cesar Reis
- Department of Preventive Medicine, Loma Linda University School of Medicine, Loma Linda, California, USA; Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Wei Yan
- Department of Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Wang
- Department of Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
28
|
Aflorei ED, Klapholz B, Chen C, Radian S, Dragu AN, Moderau N, Prodromou C, Ribeiro PS, Stanewsky R, Korbonits M. In vivo bioassay to test the pathogenicity of missense human AIP variants. J Med Genet 2018; 55:522-529. [PMID: 29632148 PMCID: PMC6073908 DOI: 10.1136/jmedgenet-2017-105191] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/23/2018] [Accepted: 03/01/2018] [Indexed: 12/17/2022]
Abstract
Background Heterozygous germline loss-of-function mutations in the aryl hydrocarbon receptor-interacting protein gene (AIP) predispose to childhood-onset pituitary tumours. The pathogenicity of missense variants may pose difficulties for genetic counselling and family follow-up. Objective To develop an in vivo system to test the pathogenicity of human AIP mutations using the fruit fly Drosophila melanogaster. Methods We generated a null mutant of the Drosophila AIP orthologue, CG1847, a gene located on the Xchromosome, which displayed lethality at larval stage in hemizygous knockout male mutants (CG1847exon1_3). We tested human missense variants of ‘unknown significance’, with ‘pathogenic’ variants as positive control. Results We found that human AIP can functionally substitute for CG1847, as heterologous overexpression of human AIP rescued male CG1847exon1_3 lethality, while a truncated version of AIP did not restore viability. Flies harbouring patient-specific missense AIP variants (p.C238Y, p.I13N, p.W73R and p.G272D) failed to rescue CG1847exon1_3 mutants, while seven variants (p.R16H, p.Q164R, p.E293V, p.A299V, p.R304Q, p.R314W and p.R325Q) showed rescue, supporting a non-pathogenic role for these latter variants corresponding to prevalence and clinical data. Conclusion Our in vivo model represents a valuable tool to characterise putative disease-causing human AIP variants and assist the genetic counselling and management of families carrying AIP variants.
Collapse
Affiliation(s)
- Elena Daniela Aflorei
- Centre for Endocrinology, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| | - Benjamin Klapholz
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Chenghao Chen
- Department of Cell and Developmental Biology, Division of Biosciences, Faculty of Life Sciences, University College London, London, UK
| | - Serban Radian
- Centre for Endocrinology, Barts and the London School of Medicine, Queen Mary University of London, London, UK.,Department of Endocrinology, C.I. Parhon National Institute of Endocrinology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Anca Neluta Dragu
- Centre for Endocrinology, Barts and the London School of Medicine, Queen Mary University of London, London, UK.,Department of Cell and Developmental Biology, Division of Biosciences, Faculty of Life Sciences, University College London, London, UK
| | - Nina Moderau
- Protein Dynamics and Cell Signalling Laboratory, Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | - Paulo S Ribeiro
- Protein Dynamics and Cell Signalling Laboratory, Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Ralf Stanewsky
- Department of Cell and Developmental Biology, Division of Biosciences, Faculty of Life Sciences, University College London, London, UK.,Institute of Neuro- and Behavioural Biology, Westfälische Wilhelms University, Münster, Germany
| | - Márta Korbonits
- Centre for Endocrinology, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| |
Collapse
|
29
|
Hernández-Ramírez LC, Morgan RM, Barry S, D’Acquisto F, Prodromou C, Korbonits M. Multi-chaperone function modulation and association with cytoskeletal proteins are key features of the function of AIP in the pituitary gland. Oncotarget 2018; 9:9177-9198. [PMID: 29507682 PMCID: PMC5823669 DOI: 10.18632/oncotarget.24183] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 01/01/2018] [Indexed: 11/25/2022] Open
Abstract
Despite the well-recognized role of loss-of-function mutations of the aryl hydrocarbon receptor interacting protein gene (AIP) predisposing to pituitary adenomas, the pituitary-specific function of this tumor suppressor remains an enigma. To determine the repertoire of interacting partners for the AIP protein in somatotroph cells, wild-type and variant AIP proteins were used for pull-down/quantitative mass spectrometry experiments against lysates of rat somatotropinoma-derived cells; relevant findings were validated by co-immunoprecipitation and co-localization. Global gene expression was studied in AIP mutation positive and negative pituitary adenomas via RNA microarrays. Direct interaction with AIP was confirmed for three known and six novel partner proteins. Novel interactions with HSPA5 and HSPA9, together with known interactions with HSP90AA1, HSP90AB1 and HSPA8, indicate that the function/stability of multiple chaperone client proteins could be perturbed by a deficient AIP co-chaperone function. Interactions with TUBB, TUBB2A, NME1 and SOD1 were also identified. The AIP variants p.R304* and p.R304Q showed impaired interactions with HSPA8, HSP90AB1, NME1 and SOD1; p.R304* also displayed reduced binding to TUBB and TUBB2A, and AIP-mutated tumors showed reduced TUBB2A expression. Our findings suggest that cytoskeletal organization, cell motility/adhesion, as well as oxidative stress responses, are functions that are likely to be involved in the tumor suppressor activity of AIP.
Collapse
Affiliation(s)
- Laura C. Hernández-Ramírez
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
- Present address: Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892-1862, USA
| | - Rhodri M.L. Morgan
- Genome Damage and Stability Centre, University of Sussex, Brighton, Falmer, BN1 9RQ, UK
- Present address: Protein Crystallography Facility, Centre for Structural Biology, Flowers Building, Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Sayka Barry
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Fulvio D’Acquisto
- Centre for Microvascular Research, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| | | | - Márta Korbonits
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
| |
Collapse
|
30
|
Cannavo S, Trimarchi F, Ferraù F. Acromegaly, genetic variants of the aryl hydrocarbon receptor pathway and environmental burden. Mol Cell Endocrinol 2017; 457:81-88. [PMID: 27998805 DOI: 10.1016/j.mce.2016.12.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 12/15/2016] [Accepted: 12/16/2016] [Indexed: 12/13/2022]
Abstract
Increasing evidence suggests that environmental contaminants can exert endocrine disruptors activities and that pollution exposition can have a role in tumorigenic processes. Several environmental pollutants have been shown to affect pituitary cells biology and function. The aryl hydrocarbon receptor (AHR) pathway is involved in xenobiotics' metabolism and in tumorigenesis. A deregulation of the AHR pathway could have a role in pituitary tumours' pathophysiology, especially in the GH secreting ones. AHR-interacting protein (AIP) is one of the key partners of AHR and is implicated in pituitary tumours' pathogenesis. Moreover, an increased prevalence of acromegaly has been reported in a highly polluted area of the province of Messina (Sicily, Italy). Nevertheless, at present, few data are available about the potential role of environmental factors in the pathogenesis and clinical expression of GH secreting pituitary tumours. This review is aimed at discussing the evidences on the potential links among environmental pollutants, the AHR pathway and the pathophysiology of GH-secreting pituitary adenomas.
Collapse
Affiliation(s)
- S Cannavo
- Department of Clinical and Experimental Medicine - Endocrinology Unit, University of Messina, Italy
| | - F Trimarchi
- Department of Clinical and Experimental Medicine - Endocrinology Unit, University of Messina, Italy
| | - F Ferraù
- Department of Clinical and Experimental Medicine - Endocrinology Unit, University of Messina, Italy.
| |
Collapse
|
31
|
Yadav RP, Artemyev NO. AIPL1: A specialized chaperone for the phototransduction effector. Cell Signal 2017; 40:183-189. [PMID: 28939106 PMCID: PMC6022367 DOI: 10.1016/j.cellsig.2017.09.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/11/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022]
Abstract
Molecular chaperones play pivotal roles in protein folding, quality control, assembly of multimeric protein complexes, protein trafficking, stress responses, and other essential cellular processes. Retinal photoreceptor rod and cone cells have an unusually high demand for production, quality control, and trafficking of key phototransduction components, and thus, require a robust and specialized chaperone machinery to ensure the fidelity of sensing and transmission of visual signals. Misfolding and/or mistrafficking of photoreceptor proteins are known causes for debilitating blinding diseases. Phosphodiesterase 6, the effector enzyme of the phototransduction cascade, relies on a unique chaperone aryl hydrocarbon receptor (AhR)-interacting protein-like 1 (AIPL1) for its stability and function. The structure of AIPL1 and its relationship with the client remained obscure until recently. This review summarizes important recent advances in understanding the mechanisms underlying normal function of AIPL1 and the protein perturbations caused by pathogenic mutations.
Collapse
Affiliation(s)
- Ravi P Yadav
- Department of Molecular Physiology and Biophysics, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States
| | - Nikolai O Artemyev
- Department of Molecular Physiology and Biophysics, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States.
| |
Collapse
|
32
|
Ibáñez-Costa A, Korbonits M. AIP and the somatostatin system in pituitary tumours. J Endocrinol 2017; 235:R101-R116. [PMID: 28835453 DOI: 10.1530/joe-17-0254] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/22/2017] [Indexed: 12/22/2022]
Abstract
Classic somatostatin analogues aimed at somatostatin receptor type 2, such as octreotide and lanreotide, represent the mainstay of medical treatment for acromegaly. These agents have the potential to decrease hormone secretion and reduce tumour size. Patients with a germline mutation in the aryl hydrocarbon receptor-interacting protein gene, AIP, develop young-onset acromegaly, poorly responsive to pharmacological therapy. In this review, we summarise the most recent studies on AIP-related pituitary adenomas, paying special attention to the causes of somatostatin resistance; the somatostatin receptor profile including type 2, type 5 and truncated variants; the role of G proteins in this pathology; the use of first and second generation somatostatin analogues; and the role of ZAC1, a zinc-finger protein with expression linked to AIP in somatotrophinoma models and acting as a key mediator of octreotide response.
Collapse
Affiliation(s)
- Alejandro Ibáñez-Costa
- Centre for EndocrinologyWilliam Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Márta Korbonits
- Centre for EndocrinologyWilliam Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| |
Collapse
|
33
|
Chen B, Liu P, Hujber EJ, Li Y, Jorgensen EM, Wang ZW. AIP limits neurotransmitter release by inhibiting calcium bursts from the ryanodine receptor. Nat Commun 2017; 8:1380. [PMID: 29123133 PMCID: PMC5680226 DOI: 10.1038/s41467-017-01704-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 10/06/2017] [Indexed: 11/16/2022] Open
Abstract
Pituitary tumors are frequently associated with mutations in the AIP gene and are sometimes associated with hypersecretion of growth hormone. It is unclear whether other factors besides an enlarged pituitary contribute to the hypersecretion. In a genetic screen for suppressors of reduced neurotransmitter release, we identified a mutation in Caenorhabditis elegans AIPR-1 (AIP-related-1), which causes profound increases in evoked and spontaneous neurotransmitter release, a high frequency of spontaneous calcium transients in motor neurons and an enlarged readily releasable pool of vesicles. Calcium bursts and hypersecretion are reversed by mutations in the ryanodine receptor but not in the voltage-gated calcium channel, indicating that these phenotypes are caused by a leaky ryanodine receptor. AIPR-1 is physically associated with the ryanodine receptor at synapses. Finally, the phenotypes in aipr-1 mutants can be rescued by presynaptic expression of mouse AIP, demonstrating that a conserved function of AIP proteins is to inhibit calcium release from ryanodine receptors.
Collapse
Affiliation(s)
- Bojun Chen
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Ping Liu
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Edward J Hujber
- Department of Biology, University of Utah, Salt Lake City, UT, 84112, USA
- Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT, 84112, USA
| | - Yan Li
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Erik M Jorgensen
- Department of Biology, University of Utah, Salt Lake City, UT, 84112, USA
- Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT, 84112, USA
| | - Zhao-Wen Wang
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA.
| |
Collapse
|
34
|
Combined x-ray crystallography and computational modeling approach to investigate the Hsp90 C-terminal peptide binding to FKBP51. Sci Rep 2017; 7:14288. [PMID: 29079741 PMCID: PMC5660230 DOI: 10.1038/s41598-017-14731-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/16/2017] [Indexed: 01/13/2023] Open
Abstract
FK506 binding protein of 51 kDa (FKBP51) is a heat shock protein 90 (Hsp90) co-chaperone involved in the regulation of steroid hormone receptors activity. It is known for its role in various regulatory pathways implicated in mood and stress-related disorders, cancer, obesity, Alzheimer’s disease and corticosteroid resistant asthma. It consists of two FKBP12 like active peptidyl prolyl isomerase (PPIase) domains (an active FK1 and inactive FK2 domain) and one tetratricopeptide repeat (TPR) domain that mediates interaction with Hsp90 via its C-terminal MEEVD peptide. Here, we report a combined x-ray crystallography and molecular dynamics study to reveal the binding mechanism of Hsp90 MEEVD peptide to the TPR domain of FKBP51. The results demonstrated that the Hsp90 C-terminal peptide binds to the TPR domain of FKBP51 with the help of di-carboxylate clamp involving Lys272, Glu273, Lys352, Asn322, and Lys329 which are conserved throughout several di-carboxylate clamp TPR proteins. Interestingly, the results from molecular dynamics study are also in agreement to the complex structure where all the contacts between these two partners were consistent throughout the simulation period. In a nutshell, our findings provide new opportunity to engage this important protein-protein interaction target by small molecules designed by structure based drug design strategy.
Collapse
|
35
|
Salvatori R, Radian S, Diekmann Y, Iacovazzo D, David A, Gabrovska P, Grassi G, Bussell AM, Stals K, Weber A, Quinton R, Crowne EC, Corazzini V, Metherell L, Kearney T, Du Plessis D, Sinha AK, Baborie A, Lecoq AL, Chanson P, Ansorge O, Ellard S, Trainer PJ, Balding D, Thomas MG, Korbonits M. In-frame seven amino-acid duplication in AIP arose over the last 3000 years, disrupts protein interaction and stability and is associated with gigantism. Eur J Endocrinol 2017; 177. [PMID: 28634279 PMCID: PMC5510572 DOI: 10.1530/eje-17-0293] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Mutations in the aryl hydrocarbon receptor-interacting protein (AIP) gene are associated with pituitary adenoma, acromegaly and gigantism. Identical alleles in unrelated pedigrees could be inherited from a common ancestor or result from recurrent mutation events. DESIGN AND METHODS Observational, inferential and experimental study, including: AIP mutation testing; reconstruction of 14 AIP-region (8.3 Mbp) haplotypes; coalescent-based approximate Bayesian estimation of the time to most recent common ancestor (tMRCA) of the derived allele; forward population simulations to estimate current number of allele carriers; proposal of mutation mechanism; protein structure predictions; co-immunoprecipitation and cycloheximide chase experiments. RESULTS Nine European-origin, unrelated c.805_825dup-positive pedigrees (four familial, five sporadic from the UK, USA and France) included 16 affected (nine gigantism/four acromegaly/two non-functioning pituitary adenoma patients and one prospectively diagnosed acromegaly patient) and nine unaffected carriers. All pedigrees shared a 2.79 Mbp haploblock around AIP with additional haploblocks privately shared between subsets of the pedigrees, indicating the existence of an evolutionarily recent common ancestor, the 'English founder', with an estimated median tMRCA of 47 generations (corresponding to 1175 years) with a confidence interval (9-113 generations, equivalent to 225-2825 years). The mutation occurred in a small tandem repeat region predisposed to slipped strand mispairing. The resulting seven amino-acid duplication disrupts interaction with HSP90 and leads to a marked reduction in protein stability. CONCLUSIONS The c.805_825dup allele, originating from a common ancestor, associates with a severe clinical phenotype and a high frequency of gigantism. The mutation is likely to be the result of slipped strand mispairing and affects protein-protein interactions and AIP protein stability.
Collapse
Affiliation(s)
| | - Serban Radian
- William Harvey Research InstituteBarts and the London School of Medicine, Queen Mary University of London, London, UK
- Department of EndocrinologyC.I. Parhon National Institute of Endocrinology, ‘C. Davila’ University of Medicine and Pharmacy, Bucharest, Romania
| | - Yoan Diekmann
- Research Department of GeneticsEvolution and Environment, University College London, London, UK
| | - Donato Iacovazzo
- William Harvey Research InstituteBarts and the London School of Medicine, Queen Mary University of London, London, UK
| | - Alessia David
- Centre of Bioinformatics and System BiologyDepartment of Life Sciences, Imperial College London, London, UK
| | - Plamena Gabrovska
- William Harvey Research InstituteBarts and the London School of Medicine, Queen Mary University of London, London, UK
| | - Giorgia Grassi
- William Harvey Research InstituteBarts and the London School of Medicine, Queen Mary University of London, London, UK
| | - Anna-Marie Bussell
- Department of Molecular GeneticsRoyal Devon and Exeter Foundation Trust, Exeter, UK
| | - Karen Stals
- Department of Molecular GeneticsRoyal Devon and Exeter Foundation Trust, Exeter, UK
| | - Astrid Weber
- Department of Clinical GeneticsLiverpool Women’s Hospital, Liverpool, UK
| | - Richard Quinton
- Department of EndocrinologyNewcastle-upon-Tyne Hospitals & Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| | - Elizabeth C Crowne
- Bristol Royal Hospital for ChildrenUniversity Hospitals Bristol Foundation Trust, Bristol, UK
| | | | - Lou Metherell
- William Harvey Research InstituteBarts and the London School of Medicine, Queen Mary University of London, London, UK
| | - Tara Kearney
- Endocrinology and Neuropathology UnitSalford Royal Hospital, Manchester, UK
| | - Daniel Du Plessis
- Endocrinology and Neuropathology UnitSalford Royal Hospital, Manchester, UK
| | | | - Atik Baborie
- The Walton Centre for Neurology and NeurosurgeryLiverpool, UK
| | - Anne-Lise Lecoq
- Assistance Publique-Hôpitaux de ParisHôpital de Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction and Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Le Kremlin-Bicêtre, France
- Inserm 1185Fac Med Paris Sud, Univ Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Philippe Chanson
- Assistance Publique-Hôpitaux de ParisHôpital de Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction and Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Le Kremlin-Bicêtre, France
- Inserm 1185Fac Med Paris Sud, Univ Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | | | - Sian Ellard
- Department of Molecular GeneticsRoyal Devon and Exeter Foundation Trust, Exeter, UK
- Institute of Biomedical and Clinical ScienceUniversity of Exeter Medical School, Exeter, UK
| | - Peter J Trainer
- Department of EndocrinologyChristie Hospital, Manchester, UK
| | - David Balding
- Research Department of GeneticsEvolution and Environment, University College London, London, UK
- Centre for Systems GenomicsSchools of Biosciences and of Mathematics & Statistics, University of Melbourne, Melbourne, Australia
| | - Mark G Thomas
- Research Department of GeneticsEvolution and Environment, University College London, London, UK
| | - Márta Korbonits
- William Harvey Research InstituteBarts and the London School of Medicine, Queen Mary University of London, London, UK
- Correspondence should be addressed to M Korbonits;
| |
Collapse
|
36
|
Scudder CJ, Niessen SJ, Catchpole B, Fowkes RC, Church DB, Forcada Y. Feline hypersomatotropism and acromegaly tumorigenesis: a potential role for the AIP gene. Domest Anim Endocrinol 2017; 59:134-139. [PMID: 28119176 DOI: 10.1016/j.domaniend.2016.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 11/14/2016] [Accepted: 11/27/2016] [Indexed: 12/15/2022]
Abstract
Acromegaly in humans is usually sporadic, however up to 20% of familial isolated pituitary adenomas are caused by germline sequence variants of the aryl-hydrocarbon-receptor interacting protein (AIP) gene. Feline acromegaly has similarities to human acromegalic families with AIP mutations. The aim of this study was to sequence the feline AIP gene, identify sequence variants and compare the AIP gene sequence between feline acromegalic and control cats, and in acromegalic siblings. The feline AIP gene was amplified through PCR using whole blood genomic DNA from 10 acromegalic and 10 control cats, and 3 sibling pairs affected by acromegaly. PCR products were sequenced and compared with the published predicted feline AIP gene. A single nonsynonymous SNP was identified in exon 1 (AIP:c.9T > G) of two acromegalic cats and none of the control cats, as well as both members of one sibling pair. The region of this SNP is considered essential for the interaction of the AIP protein with its receptor. This sequence variant has not previously been reported in humans. Two additional synonymous sequence variants were identified (AIP:c.481C > T and AIP:c.826C > T). This is the first molecular study to investigate a potential genetic cause of feline acromegaly and identified a nonsynonymous AIP single nucleotide polymorphism in 20% of the acromegalic cat population evaluated, as well as in one of the sibling pairs evaluated.
Collapse
Affiliation(s)
- C J Scudder
- Clinical Science and Services, Royal Veterinary College, Hawkshead Lane, North Mymms, Hertfordshire AL9 7TA, UK.
| | - S J Niessen
- Clinical Science and Services, Royal Veterinary College, Hawkshead Lane, North Mymms, Hertfordshire AL9 7TA, UK
| | - B Catchpole
- Department of Pathology and Pathogen Biology, Hawkshead Lane, North Mymms, Hertfordshire AL9 7TA, UK
| | - R C Fowkes
- Department of Comparative and Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - D B Church
- Clinical Science and Services, Royal Veterinary College, Hawkshead Lane, North Mymms, Hertfordshire AL9 7TA, UK
| | - Y Forcada
- Clinical Science and Services, Royal Veterinary College, Hawkshead Lane, North Mymms, Hertfordshire AL9 7TA, UK
| |
Collapse
|
37
|
Hernández-Ramírez LC, Trivellin G, Stratakis CA. Role of Phosphodiesterases on the Function of Aryl Hydrocarbon Receptor-Interacting Protein (AIP) in the Pituitary Gland and on the Evaluation of AIP Gene Variants. Horm Metab Res 2017; 49:286-295. [PMID: 28427099 DOI: 10.1055/s-0043-104700] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Familial isolated pituitary adenoma (FIPA) is caused in about 20% of cases by loss-of-function germline mutations in the AIP gene. Patients harboring AIP mutations usually present with somatotropinomas resulting either in gigantism or young-onset acromegaly. AIP encodes for a co-chaperone protein endowed with tumor suppressor properties in somatotroph cells. Among other mechanisms proposed to explain this function, a regulatory effect over the 3',5'-cyclic adenosine monophosphate (cAMP) signaling pathway seems to play a prominent role. In this setting, the well-known interaction between AIP and 2 different isoforms of phosphodiesterases (PDEs), PDE2A3 and PDE4A5, is of particular interest. While the interaction with over-expressed AIP does not seem to affect PDE2A3 function, the reported effect on PDE4A5 is, in contrast, reduced enzymatic activity. In this review, we explore the possible implications of these molecular interactions for the function of somatotroph cells. In particular, we discuss how both PDEs and AIP could act as negative regulators of the cAMP pathway in the pituitary, probably both by shared and independent mechanisms. Moreover, we describe how the evaluation of the AIP-PDE4A5 interaction has proven to be a useful tool for testing AIP mutations, complementing other in silico, in vitro, and in vivo analyses. Improved assessment of the pathogenicity of AIP mutations is indeed paramount to provide adequate guidance for genetic counseling and clinical screening in AIP mutation carriers, which can lead to prospective diagnosis of pituitary adenomas.
Collapse
Affiliation(s)
- Laura C Hernández-Ramírez
- Section on Endocrinology and Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Giampaolo Trivellin
- Section on Endocrinology and Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Constantine A Stratakis
- Section on Endocrinology and Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
38
|
Blundell KLIM, Pal M, Roe SM, Pearl LH, Prodromou C. The structure of FKBP38 in complex with the MEEVD tetratricopeptide binding-motif of Hsp90. PLoS One 2017; 12:e0173543. [PMID: 28278223 PMCID: PMC5344419 DOI: 10.1371/journal.pone.0173543] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 02/22/2017] [Indexed: 01/06/2023] Open
Abstract
Tetratricopeptide (TPR) domains are known protein interaction domains. We show that the TPR domain of FKBP8 selectively binds Hsp90, and interactions upstream of the conserved MEEVD motif are critical for tight binding. In contrast FKBP8 failed to bind intact Hsp70. The PPIase domain was not essential for the interaction with Hsp90 and binding was completely encompassed by the TPR domain alone. The conformation adopted by Hsp90 peptides, containing the conserved MEEVD motif, in the crystal structure were similar to that seen for the TPR domains of CHIP, AIP and Tah1. The carboxylate clamp interactions with bound Hsp90 peptide were a critical component of the interaction and mutation of Lys 307, involved in the carboxylate clamp, completely disrupted the interaction with Hsp90. FKBP8 binding to Hsp90 did not substantially influence its ATPase activity.
Collapse
Affiliation(s)
- Katie L. I. M. Blundell
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9RQ, England
| | - Mohinder Pal
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9RQ, England
| | - S. Mark Roe
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9RQ, England
| | - Laurence H. Pearl
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9RQ, England
| | - Chrisostomos Prodromou
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9RQ, England
- * E-mail:
| |
Collapse
|
39
|
Willhoft O, Kerr R, Patel D, Zhang W, Al-Jassar C, Daviter T, Millson SH, Thalassinos K, Vaughan CK. The crystal structure of the Sgt1-Skp1 complex: the link between Hsp90 and both SCF E3 ubiquitin ligases and kinetochores. Sci Rep 2017; 7:41626. [PMID: 28139700 PMCID: PMC5282575 DOI: 10.1038/srep41626] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/21/2016] [Indexed: 12/05/2022] Open
Abstract
The essential cochaperone Sgt1 recruits Hsp90 chaperone activity to a range of cellular factors including SCF E3 ubiquitin ligases and the kinetochore in eukaryotes. In these pathways Sgt1 interacts with Skp1, a small protein that heterodimerizes with proteins containing the F-box motif. We have determined the crystal structure of the interacting domains of Saccharomyces cerevisiae Sgt1 and Skp1 at 2.8 Å resolution and validated the interface in the context of the full-length proteins in solution. The BTB/POZ domain of Skp1 associates with Sgt1 via the concave surface of its TPR domain using residues that are conserved in humans. Dimerization of yeast Sgt1 occurs via an insertion that is absent from monomeric human Sgt1. We identify point mutations that disrupt dimerization and Skp1 binding in vitro and find that the interaction with Skp1 is an essential function of Sgt1 in yeast. Our data provide a structural rationale for understanding the phenotypes of temperature-sensitive Sgt1 mutants and for linking Skp1-associated proteins to Hsp90-dependent pathways.
Collapse
Affiliation(s)
- Oliver Willhoft
- Institute of Structural and Molecular Biology, University College London and Birkbeck, Biological Sciences, Malet Street, London, WC1E 7HX, UK
| | - Richard Kerr
- Institute of Structural and Molecular Biology, University College London and Birkbeck, Division of Biosciences, Darwin Building, Gower Street, London, WC1E 6BT, UK
| | - Dipali Patel
- Institute of Structural and Molecular Biology, University College London and Birkbeck, Biological Sciences, Malet Street, London, WC1E 7HX, UK
| | - Wenjuan Zhang
- Institute of Structural and Molecular Biology, University College London and Birkbeck, Biological Sciences, Malet Street, London, WC1E 7HX, UK
| | - Caezar Al-Jassar
- Institute of Structural and Molecular Biology, University College London and Birkbeck, Biological Sciences, Malet Street, London, WC1E 7HX, UK
| | - Tina Daviter
- Institute of Structural and Molecular Biology, University College London and Birkbeck, Biological Sciences, Malet Street, London, WC1E 7HX, UK
| | - Stefan H Millson
- School of Life Sciences, Joseph Banks Laboratory, University of Lincoln, Lincoln, LN6 7TS, UK
| | - Konstantinos Thalassinos
- Institute of Structural and Molecular Biology, University College London and Birkbeck, Division of Biosciences, Darwin Building, Gower Street, London, WC1E 6BT, UK
| | - Cara K Vaughan
- Institute of Structural and Molecular Biology, University College London and Birkbeck, Biological Sciences, Malet Street, London, WC1E 7HX, UK
| |
Collapse
|
40
|
Fukuda T, Tanaka T, Hamaguchi Y, Kawanami T, Nomiyama T, Yanase T. Augmented Growth Hormone Secretion and Stat3 Phosphorylation in an Aryl Hydrocarbon Receptor Interacting Protein (AIP)-Disrupted Somatotroph Cell Line. PLoS One 2016; 11:e0164131. [PMID: 27706259 PMCID: PMC5051713 DOI: 10.1371/journal.pone.0164131] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 08/29/2016] [Indexed: 12/22/2022] Open
Abstract
Aryl hydrocarbon receptor interacting protein (AIP) is thought to be a tumor suppressor gene, as indicated by a mutational analysis of pituitary somatotroph adenomas. However, the physiological significance of AIP inactivation in somatotroph cells remains unclear. Using CRISPR/Cas9, we identified a GH3 cell clone (termed GH3-FTY) in which Aip was genetically disrupted, and subsequently investigated its character with respect to growth hormone (Gh) synthesis and proliferation. Compared with GH3, GH3-FTY cells showed remarkably increased Gh production and a slight increase in cell proliferation. Gh-induced Stat3 phosphorylation is known to be a mechanism of Gh oversecretion in GH3. Interestingly, phosphorylated-Stat3 expression in GH3-FTY cells was increased more compared with GH3 cells, suggesting a stronger drive for this mechanism in GH3-FTY. The phenotypes of GH3-FTY concerning Gh overproduction, cell proliferation, and increased Stat3 phosphorylation were significantly reversed by the exogenous expression of Aip. GH3-FTY cells were less sensitive to somatostatin than GH3 cells in the suppression of cell proliferation, which might be associated with the reduced expression of somatostatin receptor type 2. GH3-FTY xenografts in BALB/c nude mice (GH3-FTY mice) formed more mitotic somatotroph tumors than GH3 xenografts (GH3 mice), as also evidenced by increased Ki67 scores. GH3-FTY mice were also much larger and had significantly higher plasma Gh levels than GH3 mice. Furthermore, GH3-FTY mice showed relative insulin resistance compared with GH3 mice. In conclusion, we established a somatotroph cell line, GH3-FTY, which possessed prominent Gh secretion and mitotic features associated with the disruption of Aip.
Collapse
Affiliation(s)
- Takashi Fukuda
- Department of Endocrinology and Diabetes Mellitus, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Tomoko Tanaka
- Department of Endocrinology and Diabetes Mellitus, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- Department of Bioregulatory Science of Life-related Diseases, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Yuriko Hamaguchi
- Department of Endocrinology and Diabetes Mellitus, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Takako Kawanami
- Department of Endocrinology and Diabetes Mellitus, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Takashi Nomiyama
- Department of Endocrinology and Diabetes Mellitus, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- Department of Bioregulatory Science of Life-related Diseases, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Toshihiko Yanase
- Department of Endocrinology and Diabetes Mellitus, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- Department of Bioregulatory Science of Life-related Diseases, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- * E-mail:
| |
Collapse
|
41
|
Hannah-Shmouni F, Trivellin G, Stratakis CA. Genetics of gigantism and acromegaly. Growth Horm IGF Res 2016; 30-31:37-41. [PMID: 27657986 PMCID: PMC5154831 DOI: 10.1016/j.ghir.2016.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/28/2016] [Accepted: 08/09/2016] [Indexed: 12/11/2022]
Abstract
Gigantism and acromegaly are rare disorders that are caused by excessive GH secretion and/or high levels of its mediator, IGF-1. Gigantism occurs when excess GH or IGF-1 lead to increased linear growth, before the end of puberty and epiphyseal closure. The majority of cases arise from a benign GH-secreting pituitary adenoma, with an incidence of pituitary gigantism and acromegaly of approximately 8 and 11 per million person-years, respectively. Over the past two decades, our increasing understanding of the molecular and genetic etiologies of pituitary gigantism and acromegaly yielded several genetic causes, including multiple endocrine neoplasia type 1 and 4, McCune-Albright syndrome, Carney complex, familial isolated pituitary adenoma, pituitary adenoma association due to defects in familial succinate dehydrogenase genes, and the recently identified X-linked acrogigantism. The early diagnosis of these conditions helps guide early intervention, screening, and genetic counseling of patients and their family members. In this review, we provide a concise and up-to-date discussion on the genetics of gigantism and acromegaly.
Collapse
Affiliation(s)
- Fady Hannah-Shmouni
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Giampaolo Trivellin
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Constantine A Stratakis
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
42
|
Pearl LH. Review: The HSP90 molecular chaperone-an enigmatic ATPase. Biopolymers 2016; 105:594-607. [PMID: 26991466 PMCID: PMC4879513 DOI: 10.1002/bip.22835] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/09/2016] [Accepted: 03/12/2016] [Indexed: 12/16/2022]
Abstract
The HSP90 molecular chaperone is involved in the activation and cellular stabilization of a range of 'client' proteins, of which oncogenic protein kinases and nuclear steroid hormone receptors are of particular biomedical significance. Work over the last two decades has revealed a conformational cycle critical to the biological function of HSP90, coupled to an inherent ATPase activity that is regulated and manipulated by many of the co-chaperones proteins with which it collaborates. Pharmacological inhibition of HSP90 ATPase activity results in degradation of client proteins in vivo, and is a promising target for development of new cancer therapeutics. Despite this, the actual function that HSP90s conformationally-coupled ATPase activity provides in its biological role as a molecular chaperone remains obscure. © 2016 Wiley Periodicals, Inc. Biopolymers 105: 594-607, 2016.
Collapse
Affiliation(s)
- Laurence H Pearl
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QR, UK
| |
Collapse
|
43
|
Ramírez-Rentería C, Hernández-Ramírez LC, Portocarrero-Ortiz L, Vargas G, Melgar V, Espinosa E, Espinosa-de-Los-Monteros AL, Sosa E, González B, Zúñiga S, Unterländer M, Burger J, Stals K, Bussell AM, Ellard S, Dang M, Iacovazzo D, Kapur S, Gabrovska P, Radian S, Roncaroli F, Korbonits M, Mercado M. AIP mutations in young patients with acromegaly and the Tampico Giant: the Mexican experience. Endocrine 2016; 53:402-11. [PMID: 27033541 DOI: 10.1007/s12020-016-0930-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/16/2016] [Indexed: 12/16/2022]
Abstract
Although aryl hydrocarbon receptor-interacting protein (AIP) mutations are rare in sporadic acromegaly, their prevalence among young patients is nonnegligible. The objectives of this study were to evaluate the frequency of AIP mutations in a cohort of Mexican patients with acromegaly with disease onset before the age of 30 and to search for molecular abnormalities in the AIP gene in teeth obtained from the "Tampico Giant". Peripheral blood DNA from 71 patients with acromegaly (51 females) with disease onset <30 years was analysed (median age of disease onset of 23 years) and correlated with clinical, biochemical and imaging characteristics. Sequencing was also carried out in DNA extracted from teeth of the Tampico Giant. Five patients (7 %) harboured heterozygous, germline mutations of the AIP gene. In two of them (a 9-year-old girl with gigantism and a young man with symptoms of GH excess since age 14) the c.910C>T (p.Arg304Ter), well-known truncating mutation was identified; in one of these two cases and her identical twin sister, the mutation proved to be a de novo event, since neither of their parents were found to be carriers. In the remaining three patients, new mutations were identified: a frameshift mutation (c.976_977insC, p.Gly326AfsTer), an in-frame deletion (c.872_877del, p.Val291_Leu292del) and a nonsense mutation (c.868A > T, p.Lys290Ter), which are predicted to be pathogenic based on in silico analysis. Patients with AIP mutations tended to have an earlier onset of acromegaly and harboured larger and more invasive tumours. A previously described genetic variant of unknown significance (c.869C > T, p.Ala299Val) was identified in DNA from the Tampico Giant. The prevalence of AIP mutations in young Mexican patients with acromegaly is similar to that of European cohorts. Our results support the need for genetic evaluation of patients with early onset acromegaly.
Collapse
Affiliation(s)
- Claudia Ramírez-Rentería
- Experimental Endocrinology Unit/Endocrine Service, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Sur 132 number 142, Suite 210, Colonia las Americas, Mexico City, 01120, Mexico
- Neurological Center, Centro Médico ABC, Mexico City, Mexico
| | - Laura C Hernández-Ramírez
- Centre for Endocrinology, The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | | | - Guadalupe Vargas
- Experimental Endocrinology Unit/Endocrine Service, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Sur 132 number 142, Suite 210, Colonia las Americas, Mexico City, 01120, Mexico
| | | | - Etual Espinosa
- Experimental Endocrinology Unit/Endocrine Service, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Sur 132 number 142, Suite 210, Colonia las Americas, Mexico City, 01120, Mexico
- Neurological Center, Centro Médico ABC, Mexico City, Mexico
| | - Ana Laura Espinosa-de-Los-Monteros
- Experimental Endocrinology Unit/Endocrine Service, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Sur 132 number 142, Suite 210, Colonia las Americas, Mexico City, 01120, Mexico
| | - Ernesto Sosa
- Experimental Endocrinology Unit/Endocrine Service, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Sur 132 number 142, Suite 210, Colonia las Americas, Mexico City, 01120, Mexico
| | - Baldomero González
- Experimental Endocrinology Unit/Endocrine Service, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Sur 132 number 142, Suite 210, Colonia las Americas, Mexico City, 01120, Mexico
- Neurological Center, Centro Médico ABC, Mexico City, Mexico
| | - Sergio Zúñiga
- Experimental Endocrinology Unit/Endocrine Service, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Sur 132 number 142, Suite 210, Colonia las Americas, Mexico City, 01120, Mexico
| | | | - Joachim Burger
- Institute of Anthropology, Johannes Gutenberg University, Mainz, Germany
| | - Karen Stals
- Department of Molecular Genetics, Royal Devon and Exeter, NHS Foundation Trust, Exeter, UK
| | - Anne-Marie Bussell
- Department of Molecular Genetics, Royal Devon and Exeter, NHS Foundation Trust, Exeter, UK
| | - Sian Ellard
- Department of Molecular Genetics, Royal Devon and Exeter, NHS Foundation Trust, Exeter, UK
| | - Mary Dang
- Centre for Endocrinology, The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Donato Iacovazzo
- Centre for Endocrinology, The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Sonal Kapur
- Centre for Endocrinology, The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Plamena Gabrovska
- Centre for Endocrinology, The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Serban Radian
- Centre for Endocrinology, The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Federico Roncaroli
- Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Márta Korbonits
- Centre for Endocrinology, The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Moisés Mercado
- Experimental Endocrinology Unit/Endocrine Service, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Sur 132 number 142, Suite 210, Colonia las Americas, Mexico City, 01120, Mexico.
- Neurological Center, Centro Médico ABC, Mexico City, Mexico.
| |
Collapse
|
44
|
Hernández-Ramírez LC, Martucci F, Morgan RML, Trivellin G, Tilley D, Ramos-Guajardo N, Iacovazzo D, D'Acquisto F, Prodromou C, Korbonits M. Rapid Proteasomal Degradation of Mutant Proteins Is the Primary Mechanism Leading to Tumorigenesis in Patients With Missense AIP Mutations. J Clin Endocrinol Metab 2016; 101:3144-54. [PMID: 27253664 PMCID: PMC4971335 DOI: 10.1210/jc.2016-1307] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CONTEXT The pathogenic effect of mutations in the aryl hydrocarbon receptor interacting protein (AIP) gene (AIPmuts) in pituitary adenomas is incompletely understood. We have identified the primary mechanism of loss of function for missense AIPmuts. OBJECTIVE This study sought to analyze the mechanism/speed of protein turnover of wild-type and missense AIP variants, correlating protein half-life with clinical parameters. DESIGN AND SETTING Half-life and protein-protein interaction experiments and cross-sectional analysis of AIPmut positive patients' data were performed in a clinical academic research institution. PATIENTS Data were obtained from our cohort of pituitary adenoma patients and literature-reported cases. INTERVENTIONS Protein turnover of endogenous AIP in two cell lines and fifteen AIP variants overexpressed in HEK293 cells was analyzed via cycloheximide chase and proteasome inhibition. Glutathione-S-transferase pull-down and quantitative mass spectrometry identified proteins involved in AIP degradation; results were confirmed by coimmunoprecipitation and gene knockdown. Relevant clinical data was collected. MAIN OUTCOME MEASURES Half-life of wild-type and mutant AIP proteins and its correlation with clinical parameters. RESULTS Endogenous AIP half-life was similar in HEK293 and lymphoblastoid cells (43.5 and 32.7 h). AIP variants were divided into stable proteins (median, 77.7 h; interquartile range [IQR], 60.7-92.9 h), and those with short (median, 27 h; IQR, 21.6-28.7 h) or very short (median, 7.7 h; IQR, 5.6-10.5 h) half-life; proteasomal inhibition rescued the rapid degradation of mutant proteins. The experimental half-life significantly correlated with age at diagnosis of acromegaly/gigantism (r = 0.411; P = .002). The FBXO3-containing SKP1-CUL1-F-box protein complex was identified as the E3 ubiquitin-ligase recognizing AIP. CONCLUSIONS AIP is a stable protein, driven to ubiquitination by the SKP1-CUL1-F-box protein complex. Enhanced proteasomal degradation is a novel pathogenic mechanism for AIPmuts, with direct implications for the phenotype.
Collapse
Affiliation(s)
- Laura C Hernández-Ramírez
- Centre for Endocrinology (L.C.H.-R., F.M., G.T., D.T., N.R.-G., D.I., M.K.), and Centre for Biochemical Pharmacology (F.D.), William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom; Genome Damage and Stability Centre (R.M.L.M., C.P.), University of Sussex, Brighton, Falmer, BN1 9RQ, United Kingdom
| | - Federico Martucci
- Centre for Endocrinology (L.C.H.-R., F.M., G.T., D.T., N.R.-G., D.I., M.K.), and Centre for Biochemical Pharmacology (F.D.), William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom; Genome Damage and Stability Centre (R.M.L.M., C.P.), University of Sussex, Brighton, Falmer, BN1 9RQ, United Kingdom
| | - Rhodri M L Morgan
- Centre for Endocrinology (L.C.H.-R., F.M., G.T., D.T., N.R.-G., D.I., M.K.), and Centre for Biochemical Pharmacology (F.D.), William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom; Genome Damage and Stability Centre (R.M.L.M., C.P.), University of Sussex, Brighton, Falmer, BN1 9RQ, United Kingdom
| | - Giampaolo Trivellin
- Centre for Endocrinology (L.C.H.-R., F.M., G.T., D.T., N.R.-G., D.I., M.K.), and Centre for Biochemical Pharmacology (F.D.), William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom; Genome Damage and Stability Centre (R.M.L.M., C.P.), University of Sussex, Brighton, Falmer, BN1 9RQ, United Kingdom
| | - Daniel Tilley
- Centre for Endocrinology (L.C.H.-R., F.M., G.T., D.T., N.R.-G., D.I., M.K.), and Centre for Biochemical Pharmacology (F.D.), William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom; Genome Damage and Stability Centre (R.M.L.M., C.P.), University of Sussex, Brighton, Falmer, BN1 9RQ, United Kingdom
| | - Nancy Ramos-Guajardo
- Centre for Endocrinology (L.C.H.-R., F.M., G.T., D.T., N.R.-G., D.I., M.K.), and Centre for Biochemical Pharmacology (F.D.), William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom; Genome Damage and Stability Centre (R.M.L.M., C.P.), University of Sussex, Brighton, Falmer, BN1 9RQ, United Kingdom
| | - Donato Iacovazzo
- Centre for Endocrinology (L.C.H.-R., F.M., G.T., D.T., N.R.-G., D.I., M.K.), and Centre for Biochemical Pharmacology (F.D.), William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom; Genome Damage and Stability Centre (R.M.L.M., C.P.), University of Sussex, Brighton, Falmer, BN1 9RQ, United Kingdom
| | - Fulvio D'Acquisto
- Centre for Endocrinology (L.C.H.-R., F.M., G.T., D.T., N.R.-G., D.I., M.K.), and Centre for Biochemical Pharmacology (F.D.), William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom; Genome Damage and Stability Centre (R.M.L.M., C.P.), University of Sussex, Brighton, Falmer, BN1 9RQ, United Kingdom
| | - Chrisostomos Prodromou
- Centre for Endocrinology (L.C.H.-R., F.M., G.T., D.T., N.R.-G., D.I., M.K.), and Centre for Biochemical Pharmacology (F.D.), William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom; Genome Damage and Stability Centre (R.M.L.M., C.P.), University of Sussex, Brighton, Falmer, BN1 9RQ, United Kingdom
| | - Márta Korbonits
- Centre for Endocrinology (L.C.H.-R., F.M., G.T., D.T., N.R.-G., D.I., M.K.), and Centre for Biochemical Pharmacology (F.D.), William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom; Genome Damage and Stability Centre (R.M.L.M., C.P.), University of Sussex, Brighton, Falmer, BN1 9RQ, United Kingdom
| |
Collapse
|
45
|
Signaling network map of the aryl hydrocarbon receptor. J Cell Commun Signal 2016; 10:341-346. [PMID: 27465749 DOI: 10.1007/s12079-016-0341-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 07/21/2016] [Indexed: 01/09/2023] Open
|
46
|
Neocleous V, Shammas C, Phelan MM, Fanis P, Pantelidou M, Skordis N, Mantzoros C, Phylactou LA, Toumba M. A novel MC4R deletion coexisting with FTO and MC1R gene variants, causes severe early onset obesity. Hormones (Athens) 2016; 15:441-444. [PMID: 27394708 PMCID: PMC6309374 DOI: 10.14310/horm.2002.1686] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 04/22/2016] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Heterozygous mutations on the melanocortin-4-receptor gene (MC4R) are the most frequent cause of monogenic obesity. We describe a novel MC4R deletion in a girl with severe early onset obesity, tall stature, pale skin and red hair. CASE REPORT Clinical and hormonal parameters were evaluated in a girl born full-term by non-consanguineous parents. Her body mass index (BMI) at presentation (3 years) was 30 kg/m2 (z-score: +4.5SDS). By the age of 5.2 years, she exhibited extreme linear growth acceleration and developed hyperinsulinemia. METHODS Direct sequencing of the MC4R, MC1Rand for the knownFTOsingle nucleotide polymorphism (SNP) rs9939609was performed for the patient and her family. RESULTS A novel heterozygous MC4R p.Met215del (c.643_645delATG) deletion was identified in the patient, her father and her brother, both of whom exhibited a milder phenotype. 3D structural dynamic simulation studies investigated the conformational changes induced by the p.Met215del. The patient and her mother were also found to be carriers of the obesity risk associated FTOrs9939609SNP. Finally, the identification of the known p.Arg160Trp MC1Rvariant in the patient accounts for the red hair and pale skin phenotypic features. CONCLUSION The p.Met215del causes global conformational and functional changes as it is localized at the alpha-helical transmembrane regions and the membrane spanning regions of the beta-barrel. This novel mutation produces a severe overgrowth phenotype that is apparent as from infancy and is progressive in childhood. The additional negative effect of environmental and unhealthy lifestyle habits as well as a possible co-interaction of FTOrs9939609 SNP may worsen the phenotype.
Collapse
MESH Headings
- Age of Onset
- Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism
- Body Mass Index
- Child
- Child, Preschool
- Feeding Behavior
- Female
- Gene Deletion
- Genetic Predisposition to Disease
- Hair Color/genetics
- Heredity
- Heterozygote
- Humans
- Hyperphagia/genetics
- Hyperphagia/physiopathology
- Pediatric Obesity/diagnosis
- Pediatric Obesity/genetics
- Pediatric Obesity/physiopathology
- Pedigree
- Phenotype
- Polymorphism, Single Nucleotide
- Protein Conformation
- Receptor, Melanocortin, Type 1/genetics
- Receptor, Melanocortin, Type 4/chemistry
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/metabolism
- Severity of Illness Index
- Skin Pigmentation/genetics
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Vassos Neocleous
- Department of Molecular Genetics, Function & Therapy, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - Christos Shammas
- Department of Molecular Genetics, Function & Therapy, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - Marie M Phelan
- Department of Biochemistry NMR Centre for Structural Biology, University of Liverpool, Institute of Integrative Biology, Crown Street, Liverpool, UK
| | - Pavlos Fanis
- Department of Molecular Genetics, Function & Therapy, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - Maria Pantelidou
- Department of Pharmacy and Department of Nursing, School of Health Sciences, Frederick University, Nicosia, Cyprus
| | - Nicos Skordis
- Department of Molecular Genetics, Function & Therapy, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
- Division of Pediatric Endocrinology, Paedi Center for specialized Pediatrics, Nicosia, Cyprus
- St George's University of London Medical School at the University of Nicosia, Nicosia, Cyprus
| | - Christos Mantzoros
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, USA
| | - Leonidas A Phylactou
- Department of Molecular Genetics, Function & Therapy, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - Meropi Toumba
- Department of Molecular Genetics, Function & Therapy, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus.
- Paediatrics & Paediatric Endocrinology Clinic, IASIS Hospital, 8036, Paphos, Cyprus.
| |
Collapse
|
47
|
Gopalakrishna KN, Boyd K, Yadav RP, Artemyev NO. Aryl Hydrocarbon Receptor-interacting Protein-like 1 Is an Obligate Chaperone of Phosphodiesterase 6 and Is Assisted by the γ-Subunit of Its Client. J Biol Chem 2016; 291:16282-91. [PMID: 27268253 DOI: 10.1074/jbc.m116.737593] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Indexed: 12/26/2022] Open
Abstract
Phosphodiesterase 6 (PDE6) is the effector enzyme in the phototransduction cascade and is critical for the health of both rod and cone photoreceptors. Its dysfunction, caused by mutations in either the enzyme itself or AIPL1 (aryl hydrocarbon receptor-interacting protein-like 1), leads to retinal diseases culminating in blindness. Progress in research on PDE6 and AIPL1 has been severely hampered by failure to express functional PDE6 in a heterologous expression system. Here, we demonstrated that AIPL1 is an obligate chaperone of PDE6 and that it enables low yield functional folding of cone PDE6C in cultured cells. We further show that the AIPL1-mediated production of folded PDE6C is markedly elevated in the presence of the inhibitory Pγ-subunit of PDE6. As illustrated in this study, a simple and sensitive system in which AIPL1 and Pγ are co-expressed with PDE6 represents an effective tool for probing structure-function relationships of AIPL1 and reliably establishing the pathogenicity of its variants.
Collapse
Affiliation(s)
| | - Kimberly Boyd
- From the Departments of Molecular Physiology and Biophysics and
| | - Ravi P Yadav
- From the Departments of Molecular Physiology and Biophysics and
| | - Nikolai O Artemyev
- From the Departments of Molecular Physiology and Biophysics and Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| |
Collapse
|
48
|
Bolger GB, Bizzi MF, Pinheiro SV, Trivellin G, Smoot L, Accavitti MA, Korbonits M, Ribeiro-Oliveira A. cAMP-specific PDE4 phosphodiesterases and AIP in the pathogenesis of pituitary tumors. Endocr Relat Cancer 2016; 23:419-31. [PMID: 27267386 PMCID: PMC4901527 DOI: 10.1530/erc-15-0205] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 04/18/2016] [Indexed: 12/27/2022]
Abstract
PDE4 cyclic nucleotide phosphodiesterases regulate cAMP abundance in cells and therefore regulate numerous processes, including cell growth and differentiation. The rat PDE4A5 isoform (human homolog PDE4A4) interacts with the AIP protein (also called XAP2 or ARA-9). Germline mutations in AIP occur in approximately 20% of patients with Familial Isolated Pituitary Adenoma (FIPA) and 20% of childhood-onset simplex somatotroph adenomas. We therefore examined the protein expression of PDE4A4 and the closely related isoform PDE4A8 in normal human pituitary tissue and in pituitary adenomas. PDE4A4 had low expression in normal pituitary but was significantly overexpressed in somatotroph, lactotroph, corticotroph and clinically nonfunctioning gonadotroph adenomas (P<0.0001 for all subtypes). Likewise, PDE4A8 was expressed in normal pituitary and was also significantly overexpressed in the adenoma subtypes (P<0.0001 for all). Among the different adenoma subtypes, corticotroph and lactotroph adenomas were the highest and lowest expressed for PDE4A4, respectively, whereas the opposite was observed for PDE4A8. Naturally occurring oncogenic variants in AIP were shown by a two-hybrid assay to disrupt the ability of AIP to interact with PDE4A5. A reverse two-hybrid screen identified numerous additional variants in the tetratricopeptide repeat (TPR) region of AIP that also disrupted its ability to interact with PDE4A5. The expression of PDE4A4 and PDE4A8 in normal pituitary, their increased expression in adenomatous pituitary cells where AIP is meant to participate, and the disruption of the PDE4A4-AIP interaction by AIP mutants may play a role in pituitary tumorigenesis.
Collapse
Affiliation(s)
- Graeme B Bolger
- Department of MedicineUniversity of Alabama at Birmingham, Birmingham, Alabama, USA Department of PharmacologyUniversity of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mariana F Bizzi
- Department of Internal MedicineFederal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio V Pinheiro
- Department of PediatricsFederal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Giampaolo Trivellin
- Center for EndocrinologyWilliam Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Lisa Smoot
- Department of MedicineUniversity of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mary-Ann Accavitti
- Department of Microbiology and ImmunologyUniversity of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Márta Korbonits
- Center for EndocrinologyWilliam Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Antonio Ribeiro-Oliveira
- Department of Internal MedicineFederal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
49
|
Lecoq AL, Viengchareun S, Hage M, Bouligand J, Young J, Boutron A, Zizzari P, Lombès M, Chanson P, Kamenický P. AIP mutations impair AhR signaling in pituitary adenoma patients fibroblasts and in GH3 cells. Endocr Relat Cancer 2016; 23:433-43. [PMID: 27080473 DOI: 10.1530/erc-16-0041] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 04/14/2016] [Indexed: 12/30/2022]
Abstract
Germline mutations in the aryl hydrocarbon receptor-interacting protein (AIP) gene predispose humans to pituitary adenomas through unknown molecular mechanisms. The best-known interacting partner of AIP is the aryl hydrocarbon receptor (AhR), a transcription factor that mediates the effects of xenobiotics implicated in carcinogenesis. As 75% of AIP mutations disrupt the physical and/or functional interaction with AhR, we postulated that the tumorigenic potential of AIP mutations might result from altered AhR signaling. We evaluated the impact of AIP mutations on the AhR signaling pathway, first in fibroblasts from AIP-mutated patients with pituitary adenomas, by comparison with fibroblasts from healthy subjects, then in transfected pituitary GH3 cells. The AIP protein level in mutated fibroblasts was about half of that in cells from healthy subjects, but AhR expression was unaffected. Gene expression analyses showed significant modifications in the expression of the AhR target genes CYP1B1 and AHRR in AIP-mutated fibroblasts, both before and after stimulation with the endogenous AhR ligand kynurenine. Kynurenine increased Cyp1b1 expression to a greater extent in GH3 cells overexpressing wild type compared with cells expressing mutant AIP Knockdown of endogenous Aip in these cells attenuated Cyp1b1 induction by the AhR ligand. Both mutant AIP expression and knockdown of endogenous Aip affected the kynurenine-dependent GH secretion of GH3 cells. This study of human fibroblasts bearing endogenous heterozygous AIP mutations and transfected pituitary GH3 cells shows that AIP mutations affect the AIP protein level and alter AhR transcriptional activity in a gene- and tissue-dependent manner.
Collapse
Affiliation(s)
- Anne-Lise Lecoq
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1185Le Kremlin-Bicêtre, France Université Paris-SudFaculté de Médecine Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Say Viengchareun
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1185Le Kremlin-Bicêtre, France Université Paris-SudFaculté de Médecine Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Mirella Hage
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1185Le Kremlin-Bicêtre, France Université Paris-SudFaculté de Médecine Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Jérôme Bouligand
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1185Le Kremlin-Bicêtre, France Université Paris-SudFaculté de Médecine Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France Assistance Publique-Hôpitaux de ParisService de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Jacques Young
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1185Le Kremlin-Bicêtre, France Université Paris-SudFaculté de Médecine Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France Assistance Publique-Hôpitaux de ParisService d'Endocrinologie et des Maladies de la Reproduction, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Audrey Boutron
- Assistance Publique-Hôpitaux de ParisService de Biochimie, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Philippe Zizzari
- Inserm U894Centre de Psychiatrie et Neurosciences, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marc Lombès
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1185Le Kremlin-Bicêtre, France Université Paris-SudFaculté de Médecine Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France Assistance Publique-Hôpitaux de ParisService d'Endocrinologie et des Maladies de la Reproduction, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Philippe Chanson
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1185Le Kremlin-Bicêtre, France Université Paris-SudFaculté de Médecine Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France Assistance Publique-Hôpitaux de ParisService d'Endocrinologie et des Maladies de la Reproduction, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Peter Kamenický
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1185Le Kremlin-Bicêtre, France Université Paris-SudFaculté de Médecine Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, France Assistance Publique-Hôpitaux de ParisService d'Endocrinologie et des Maladies de la Reproduction, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| |
Collapse
|
50
|
Yadav RP, Majumder A, Gakhar L, Artemyev NO. Extended conformation of the proline-rich domain of human aryl hydrocarbon receptor-interacting protein-like 1: implications for retina disease. J Neurochem 2015; 135:165-75. [PMID: 26139345 DOI: 10.1111/jnc.13223] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 05/29/2015] [Accepted: 06/25/2015] [Indexed: 12/18/2022]
Abstract
Mutations in the primate-specific proline-rich domain (PRD) of aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) are thought to cause Leber congenital amaurosis or dominant cone-rod dystrophy. The role of PRD and the mechanisms of PRD mutations are poorly understood. Here, we have examined properties of hAIPL1 and effects of the PRD mutations on protein structure and function. Solution structures of hAIPL1, hAIPL11-316 with PRD truncation, and the P351Δ12 and P376S mutants were examined by small angle X-ray scattering. Our analysis suggests that PRD assumes an extended conformation and does not interact with the FK506-binding and tetratricopeptide domains. The PRD truncation, but not PRD mutations, reduced the molecule's radius of gyration and maximum dimension. We demonstrate that hAIPL1 is a monomeric protein, and its secondary structure and stability are not affected by the PRD mutations. PRD itself is an extended monomeric random coil. The PRD mutations caused little or no changes in hAIPL1 binding to known partners, phosphodiesterase-6A and HSP90. We also identified the γ-subunit of phosphodiesterase-6 as a novel partner of hAIPL1 and hypothesize that this interaction is altered by P351Δ12. Our results highlight the complexity of mechanisms of PRD mutations in disease and the possibility that certain mutations are benign variants. Mutations in the proline-rich domain (PRD) of human AIPL1 cause severe retinal diseases, yet the role of PRD and the mechanisms of PRD mutations are unknown. Here, we describe a SAXS-derived solution structure of AIPL1 and functional properties of disease-linked AIPL1-PRD mutants. This structure and functional analyses provide a framework for understanding the mechanisms of PRD in disease.
Collapse
Affiliation(s)
- Ravi P Yadav
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, USA
| | - Anurima Majumder
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, USA
| | - Lokesh Gakhar
- Department of Biochemistry, University of Iowa, Iowa City, Iowa, USA.,Protein Crystallography Facility, University of Iowa, Iowa City, Iowa, USA
| | - Nikolai O Artemyev
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|