1
|
Jang HB, Lee DH, Jung S, Lim SC. Safety of Simultaneous Surgery for Pituitary Neoplasms and Rhinosinusitis. Clin Otolaryngol 2024. [PMID: 39483087 DOI: 10.1111/coa.14248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 07/25/2024] [Accepted: 10/13/2024] [Indexed: 11/03/2024]
Affiliation(s)
- Hye-Bin Jang
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School & Hwasun Hospital, Hwasun, South Korea
| | - Dong Hoon Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School & Hwasun Hospital, Hwasun, South Korea
| | - Shin Jung
- Department of Neurosurgery, Chonnam National University Medical School & Hwasun Hospital, Hwasun, South Korea
| | - Sang Chul Lim
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School & Hwasun Hospital, Hwasun, South Korea
| |
Collapse
|
2
|
Hosea R, Hillary S, Naqvi S, Wu S, Kasim V. The two sides of chromosomal instability: drivers and brakes in cancer. Signal Transduct Target Ther 2024; 9:75. [PMID: 38553459 PMCID: PMC10980778 DOI: 10.1038/s41392-024-01767-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/18/2024] [Accepted: 02/06/2024] [Indexed: 04/02/2024] Open
Abstract
Chromosomal instability (CIN) is a hallmark of cancer and is associated with tumor cell malignancy. CIN triggers a chain reaction in cells leading to chromosomal abnormalities, including deviations from the normal chromosome number or structural changes in chromosomes. CIN arises from errors in DNA replication and chromosome segregation during cell division, leading to the formation of cells with abnormal number and/or structure of chromosomes. Errors in DNA replication result from abnormal replication licensing as well as replication stress, such as double-strand breaks and stalled replication forks; meanwhile, errors in chromosome segregation stem from defects in chromosome segregation machinery, including centrosome amplification, erroneous microtubule-kinetochore attachments, spindle assembly checkpoint, or defective sister chromatids cohesion. In normal cells, CIN is deleterious and is associated with DNA damage, proteotoxic stress, metabolic alteration, cell cycle arrest, and senescence. Paradoxically, despite these negative consequences, CIN is one of the hallmarks of cancer found in over 90% of solid tumors and in blood cancers. Furthermore, CIN could endow tumors with enhanced adaptation capabilities due to increased intratumor heterogeneity, thereby facilitating adaptive resistance to therapies; however, excessive CIN could induce tumor cells death, leading to the "just-right" model for CIN in tumors. Elucidating the complex nature of CIN is crucial for understanding the dynamics of tumorigenesis and for developing effective anti-tumor treatments. This review provides an overview of causes and consequences of CIN, as well as the paradox of CIN, a phenomenon that continues to perplex researchers. Finally, this review explores the potential of CIN-based anti-tumor therapy.
Collapse
Affiliation(s)
- Rendy Hosea
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Sharon Hillary
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Sumera Naqvi
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China.
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China.
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
3
|
Sorensen Turpin CG, Sloan D, LaForest M, Klebanow LU, Mitchell D, Severson AF, Bembenek JN. Securin Regulates the Spatiotemporal Dynamics of Separase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.571338. [PMID: 38168402 PMCID: PMC10760073 DOI: 10.1101/2023.12.12.571338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Separase is a key regulator of the metaphase to anaphase transition with multiple functions. Separase cleaves cohesin to allow chromosome segregation and localizes to vesicles to promote exocytosis in mid-anaphase. The anaphase promoting complex/cyclosome (APC/C) activates separase by ubiquitinating its inhibitory chaperone, securin, triggering its degradation. How this pathway controls the exocytic function of separase has not been investigated. During meiosis I, securin is degraded over several minutes, while separase rapidly relocalizes from kinetochore structures at the spindle and cortex to sites of action on chromosomes and vesicles at anaphase onset. The loss of cohesin coincides with the relocalization of separase to the chromosome midbivalent at anaphase onset. APC/C depletion prevents separase relocalization, while securin depletion causes precocious separase relocalization. Expression of non-degradable securin inhibits chromosome segregation, exocytosis, and separase localization to vesicles but not to the anaphase spindle. We conclude that APC/C mediated securin degradation controls separase localization. This spatiotemporal regulation will impact the effective local concentration of separase for more precise targeting of substrates in anaphase.
Collapse
Affiliation(s)
- Christopher G. Sorensen Turpin
- Current Address: Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Dillon Sloan
- Current Address: Department of Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Marian LaForest
- Current Address: Columbia University, Herbert Irving Comprehensive Cancer Center, NYC, New York, United States of America
| | | | - Diana Mitchell
- Current Address: Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Aaron F. Severson
- Current Address: Center for Gene Regulation in Health and Disease and Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, Ohio, United States of America
| | - Joshua N. Bembenek
- Current Address: Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| |
Collapse
|
4
|
Preclinical Models of Neuroendocrine Neoplasia. Cancers (Basel) 2022; 14:cancers14225646. [PMID: 36428741 PMCID: PMC9688518 DOI: 10.3390/cancers14225646] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
Neuroendocrine neoplasia (NENs) are a complex and heterogeneous group of cancers that can arise from neuroendocrine tissues throughout the body and differentiate them from other tumors. Their low incidence and high diversity make many of them orphan conditions characterized by a low incidence and few dedicated clinical trials. Study of the molecular and genetic nature of these diseases is limited in comparison to more common cancers and more dependent on preclinical models, including both in vitro models (such as cell lines and 3D models) and in vivo models (such as patient derived xenografts (PDXs) and genetically-engineered mouse models (GEMMs)). While preclinical models do not fully recapitulate the nature of these cancers in patients, they are useful tools in investigation of the basic biology and early-stage investigation for evaluation of treatments for these cancers. We review available preclinical models for each type of NEN and discuss their history as well as their current use and translation.
Collapse
|
5
|
Huang J, Zhang F, Hu G, Pan Y, Sun W, Jiang L, Wang P, Qiu J, Ding X. SIRT1 suppresses pituitary tumor progression by downregulating PTTG1 expression. Oncol Rep 2022; 48:143. [PMID: 35730625 DOI: 10.3892/or.2022.8354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/03/2022] [Indexed: 11/06/2022] Open
Abstract
Although pituitary tumors are among the most common types of brain tumor, the underlying molecular mechanism of this disease remains obscure. To this end, the role of sirtuin 1 (SIRT1) in pituitary tumors was reported. The results of reverse transcription‑quantitative PCR and immunohistochemistry revealed that sirtuin 1 (SIRT1) expression was downregulated in the tumor tissues of patients with pituitary tumors. In vitro experiments of the present study demonstrated that SIRT1 upregulation suppressed pituitary tumor cell line growth, while SIRT1 downregulation demonstrated the opposite effect. Additionally, it was determined that the enzymatic activity of SIRT1 was required for its cellular function. A mechanistic experiment determined that SIRT1 negatively regulated pituitary tumor‑transforming gene 1 (PTTG1) expression through the deacetylation of histone (H)3 lysine (K)9ac at the promoter region of PTTG1. Moreover, H3K9ac levels at the PTTG1 promoter were determined to be an essential regulatory element for PTTG1 expression. Thus, it was concluded that the SIRT1/H3K9ac/PTTG1 axis contributed to pituitary tumor formation and may represent a potential therapeutic strategy.
Collapse
Affiliation(s)
- Jinxiang Huang
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Fenglin Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Guohan Hu
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Yuan Pan
- Department of Neurosurgery, No. 971 Hospital of People's Liberation Army Navy, Qingdao, Shandong 266071, P.R. China
| | - Wei Sun
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Lei Jiang
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Peng Wang
- Department of Radiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Jiting Qiu
- Department of Neurosurgery, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai 201803, P.R. China
| | - Xuehua Ding
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
6
|
Välimäki N, Schalin-Jäntti C, Karppinen A, Paetau A, Kivipelto L, Aaltonen LA, Karhu A. Genetic and Epigenetic Characterization of Growth Hormone-Secreting Pituitary Tumors. Mol Cancer Res 2019; 17:2432-2443. [PMID: 31578227 DOI: 10.1158/1541-7786.mcr-19-0434] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/25/2019] [Accepted: 09/27/2019] [Indexed: 11/16/2022]
Abstract
Somatic driver mechanisms of pituitary adenoma pathogenesis have remained incompletely characterized; apart from mutations in the stimulatory Gα protein (Gαs encoded by GNAS) causing activated cAMP synthesis, pathogenic variants are rarely found in growth hormone-secreting pituitary tumors (somatotropinomas). The purpose of the current work was to clarify how genetic and epigenetic alterations contribute to the development of somatotropinomas by conducting an integrated copy number alteration, whole-genome and bisulfite sequencing, and transcriptome analysis of 21 tumors. Somatic mutation burden was low, but somatotropinomas formed two subtypes associated with distinct aneuploidy rates and unique transcription profiles. Tumors with recurrent chromosome aneuploidy (CA) were GNAS mutation negative (Gsp- ). The chromosome stable (CS) -group contained Gsp+ somatotropinomas and two totally aneuploidy-free Gsp- tumors. Genes related to the mitotic G1-S-checkpoint transition were differentially expressed in CA- and CS-tumors, indicating difference in mitotic progression. Also, pituitary tumor transforming gene 1 (PTTG1), a regulator of sister chromatid segregation, showed abundant expression in CA-tumors. Moreover, somatotropinomas displayed distinct Gsp genotype-specific methylation profiles and expression quantitative methylation (eQTM) analysis revealed that inhibitory Gα (Gαi) signaling is activated in Gsp+ tumors. These findings suggest that aneuploidy through modulated driver pathways may be a causative mechanism for tumorigenesis in Gsp- somatotropinomas, whereas Gsp+ tumors with constitutively activated cAMP synthesis seem to be characterized by DNA methylation activated Gαi signaling. IMPLICATIONS: These findings provide valuable new information about subtype-specific pituitary tumorigenesis and may help to elucidate the mechanisms of aneuploidy also in other tumor types.
Collapse
Affiliation(s)
- Niko Välimäki
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland.,Applied Tumor Genomics, Research Programs Unit, FI-00014 University of Helsinki, Finland
| | - Camilla Schalin-Jäntti
- Endocrinology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Atte Karppinen
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anders Paetau
- Department of Pathology, HUSLAB, University of Helsinki, Helsinki, Finland
| | - Leena Kivipelto
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Lauri A Aaltonen
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland.,Applied Tumor Genomics, Research Programs Unit, FI-00014 University of Helsinki, Finland
| | - Auli Karhu
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland. .,Applied Tumor Genomics, Research Programs Unit, FI-00014 University of Helsinki, Finland
| |
Collapse
|
7
|
Xekouki P, Brennand A, Whitelaw B, Pacak K, Stratakis CA. The 3PAs: An Update on the Association of Pheochromocytomas, Paragangliomas, and Pituitary Tumors. Horm Metab Res 2019; 51:419-436. [PMID: 30273935 PMCID: PMC7448524 DOI: 10.1055/a-0661-0341] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pituitary adenomas (PA) and pheochromocytomas/paragangliomas (PHEO/PGL) are rare tumors. Although they may co-exist by coincidence, there is mounting evidence that genes predisposing in PHEO/PGL development, may play a role in pituitary tumorigenesis. In 2012, we described a GH-secreting PA caused by an SDHD mutation in a patient with familial PGLs and found loss of heterozygosity at the SDHD locus in the pituitary tumor, along with increased hypoxia-inducible factor 1α (HIF-1α) levels. Additional patients with PAs and SDHx defects have since been reported. Overall, prevalence of SDHx mutations in PA is very rare (0.3-1.8% in unselected cases) but we and others have identified several cases of PAs with PHEOs/PGLs, like our original report, a condition which we termed the 3 P association (3PAs). Interestingly, when 3PAs is found in the sporadic setting, no SDHx defects were identified, whereas in familial PGLs, SDHx mutations were identified in 62.5-75% of the reported cases. Hence, pituitary surveillance is recommended among patients with SDHx defects. It is possible that the SDHx germline mutation-negative 3PAs cases may be due to another gene, epigenetic changes, mutations in modifier genes, mosaicism, somatic mutations, pituitary hyperplasia due to ectopic hypothalamic hormone secretion or a coincidence. PA in 3PAs are mainly macroadenomas, more aggressive, more resistant to somatostatin analogues, and often require surgery. Using the Sdhb +/- mouse model, we showed that hyperplasia may be the first abnormality in tumorigenesis as initial response to pseudohypoxia. We also propose surveillance and follow-up approach of patients presenting with this association.
Collapse
Affiliation(s)
- Paraskevi Xekouki
- Department of Endocrinology, King’s College Hospital, London, UK
- Division of Diabetes & Nutritional Sciences, King’s College London, London, UK
| | - Ana Brennand
- Division of Diabetes & Nutritional Sciences, King’s College London, London, UK
| | - Ben Whitelaw
- Department of Endocrinology, King’s College Hospital, London, UK
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Constantine A. Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| |
Collapse
|
8
|
Gahete MD, Jimenez-Vacas JM, Alors-Perez E, Herrero-Aguayo V, Fuentes-Fayos AC, Pedraza-Arevalo S, Castaño JP, Luque RM. Mouse models in endocrine tumors. J Endocrinol 2018; 240:JOE-18-0571.R1. [PMID: 30475226 DOI: 10.1530/joe-18-0571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022]
Abstract
Endocrine and neuroendocrine tumors comprise a highly heterogeneous group of neoplasms that can arise from (neuro)endocrine cells, either from endocrine glands or from the widespread diffuse neuroendocrine system, and, consequently, are widely distributed throughout the body. Due to their diversity, heterogeneity and limited incidence, studying in detail the molecular and genetic alterations that underlie their development and progression is still a highly elusive task. This, in turn, hinders the discovery of novel therapeutic options for these tumors. To circumvent these limitations, numerous mouse models of endocrine and neuroendocrine tumors have been developed, characterized and used in pre-clinical, co-clinical (implemented in mouse models and patients simultaneously) and post-clinical studies, for they represent powerful and necessary tools in basic and translational tumor biology research. Indeed, different in vivo mouse models, including cell line-based xenografts (CDXs), patient-derived xenografts (PDXs) and genetically engineered mouse models (GEMs), have been used to delineate the development, progression and behavior of human tumors. Results gained with these in vivo models have facilitated the clinical application in patients of diverse breakthrough discoveries made in this field. Herein, we review the generation, characterization and translatability of the most prominent mouse models of endocrine and neuroendocrine tumors reported to date, as well as the most relevant clinical implications obtained for each endocrine and neuroendocrine tumor type.
Collapse
Affiliation(s)
- Manuel D Gahete
- M Gahete, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, 14011, Spain
| | - Juan M Jimenez-Vacas
- J Jimenez-Vacas, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Emilia Alors-Perez
- E Alors-Perez, Department of Cell Biology, Physiology and Inmunology, Maimonides Institute for Biomedical Research of Cordoba (IMIBIC) / University of Cordoba, Cordoba, Spain
| | - Vicente Herrero-Aguayo
- V Herrero-Aguayo, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- A Fuentes-Fayos, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Sergio Pedraza-Arevalo
- S Pedraza-Arevalo, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Justo P Castaño
- J Castaño, Dpt. of Cell Biology-University of Córdoba, IMIBIC-Maimonides Biomedical Research Institute of Cordoba, Cordoba, E-14004, Spain
| | - Raul M Luque
- R Luque, Dept of Cell Biology, Phisiology and Inmunology, Section of Cell Biology, University of Cordoba, Cordoba, Spain, Cordoba, 14014, Spain
| |
Collapse
|
9
|
Fedele M, Paciello O, De Biase D, Monaco M, Chiappetta G, Vitiello M, Barbieri A, Rea D, Luciano A, Papparella S, Arra C, Fusco A. HMGA2 cooperates with either p27 kip1 deficiency or Cdk4 R24C mutation in pituitary tumorigenesis. Cell Cycle 2018; 17:580-588. [PMID: 29157111 DOI: 10.1080/15384101.2017.1403682] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We have previously reported a critical role of HMGA proteins in pituitary tumorigenesis since either the Hmga1 or Hmga2 gene overexpression/activation induces the development of mixed growth hormone/prolactin cell pituitary adenomas by activating the E2F transcription factor 1, and then enhancing the G1/S transition of the cell cycle. Consistently, amplification and overexpression of the HMGA2 gene was found in human pituitary prolactinomas. Since impairment of the cell cycle control represents a feature of experimental and human pituitary adenomas, we have investigated the possible synergism between the alterations of other cell cycle regulators, such as p27 deficiency or Cdk4R24C mutation, with Hmga2 overexpression in pituitary tumorigenesis. Therefore, we crossed the Hmga2/T mice, overexpressing the truncated/active form of the Hmga2 gene, either with the knockout mice for p27kip1, or with the knockin mice for the Cdk4R24C mutation, both developing pituitary adenomas. Increased incidence and decreased latency in the development of pituitary lesions appeared in double mutant Hmga2/T;Cdk4R24C mice, and increased features of invasiveness and atypia were observed in pituitary tumors of both Hmga2/T;p27-ko and Hmga2/T;Cdk4R24C double mutant mice as compared with single mutant compounds. Interestingly, most of these mice develop pituitary adenomas with high Ki67 index, extrasellar expansion and brain tissue infiltration, representing good mouse models for human aggressive pituitary adenomas. Taken together, the results reported here indicate a cooperation between HMGA2 overexpression and either p27kip1 or CDK4 impairment in promoting pituitary tumor development and progression.
Collapse
Affiliation(s)
- Monica Fedele
- a CNR - Institute of Experimental Endocrinology and Oncology - c/o Department of Molecular Medicine and Medical Biotechnologies , University of Naples "Federico II" , Naples , Italy
| | - Orlando Paciello
- b Department of Veterinary Medicine and animal production , University of Naples "Federico II" , Naples , Italy
| | - Davide De Biase
- b Department of Veterinary Medicine and animal production , University of Naples "Federico II" , Naples , Italy
| | - Mario Monaco
- c Dipartimento di Ricerca Traslazionale a supporto dei percorsi oncologici, S.C. Genomica Funzionale e S.S.D. Sperimentazione Animale , Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale , Naples , Italy
| | - Gennaro Chiappetta
- c Dipartimento di Ricerca Traslazionale a supporto dei percorsi oncologici, S.C. Genomica Funzionale e S.S.D. Sperimentazione Animale , Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale , Naples , Italy
| | - Michela Vitiello
- a CNR - Institute of Experimental Endocrinology and Oncology - c/o Department of Molecular Medicine and Medical Biotechnologies , University of Naples "Federico II" , Naples , Italy
| | - Antonio Barbieri
- c Dipartimento di Ricerca Traslazionale a supporto dei percorsi oncologici, S.C. Genomica Funzionale e S.S.D. Sperimentazione Animale , Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale , Naples , Italy
| | - Domenica Rea
- c Dipartimento di Ricerca Traslazionale a supporto dei percorsi oncologici, S.C. Genomica Funzionale e S.S.D. Sperimentazione Animale , Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale , Naples , Italy
| | - Antonio Luciano
- c Dipartimento di Ricerca Traslazionale a supporto dei percorsi oncologici, S.C. Genomica Funzionale e S.S.D. Sperimentazione Animale , Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale , Naples , Italy
| | - Serenella Papparella
- b Department of Veterinary Medicine and animal production , University of Naples "Federico II" , Naples , Italy
| | - Claudio Arra
- c Dipartimento di Ricerca Traslazionale a supporto dei percorsi oncologici, S.C. Genomica Funzionale e S.S.D. Sperimentazione Animale , Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale , Naples , Italy
| | - Alfredo Fusco
- a CNR - Institute of Experimental Endocrinology and Oncology - c/o Department of Molecular Medicine and Medical Biotechnologies , University of Naples "Federico II" , Naples , Italy
| |
Collapse
|
10
|
Sapochnik M, Fuertes M, Arzt E. Programmed cell senescence: role of IL-6 in the pituitary. J Mol Endocrinol 2017; 58:R241-R253. [PMID: 28381401 DOI: 10.1530/jme-17-0026] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/05/2017] [Indexed: 12/11/2022]
Abstract
IL-6 is a pleiotropic cytokine with multiple pathophysiological functions. As a key factor of the senescence secretome, it can not only promote tumorigenesis and cell proliferation but also exert tumor suppressive functions, depending on the cellular context. IL-6, as do other cytokines, plays important roles in the function, growth and neuroendocrine responses of the anterior pituitary gland. The multiple actions of IL-6 on normal and adenomatous pituitary function, cell proliferation, angiogenesis and extracellular matrix remodeling indicate its importance in the regulation of the anterior pituitary. Pituitary tumors are mostly benign adenomas with low mitotic index and rarely became malignant. Premature senescence occurs in slow-growing benign tumors, like pituitary adenomas. The dual role of IL-6 in senescence and tumorigenesis is well represented in pituitary tumor development, as it has been demonstrated that effects of paracrine IL-6 may allow initial pituitary cell growth, whereas autocrine IL-6 in the same tumor triggers senescence and restrains aggressive growth and malignant transformation. IL-6 is instrumental in promotion and maintenance of the senescence program in pituitary adenomas.
Collapse
Affiliation(s)
- Melanie Sapochnik
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - Mariana Fuertes
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - Eduardo Arzt
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
- Departamento de Fisiología y Biología Molecular y CelularFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
11
|
mTOR promotes pituitary tumor development through activation of PTTG1. Oncogene 2016; 36:979-988. [PMID: 27524416 DOI: 10.1038/onc.2016.264] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 05/19/2016] [Accepted: 06/16/2016] [Indexed: 12/11/2022]
Abstract
As one of the most common intracranial tumors, pituitary tumor is associated with high morbidity. Effective therapy is currently not available for some pituitary tumors due to the largely undefined pathological processes of pituitary tumorigenesis. In this study, hyperactivation of mammalian/mechanistic target of rapamycin (mTOR) signaling was observed in estrogen-induced rat pituitary tumor and mTOR inhibitor rapamycin blocked the tumor development. Pituitary knockout of either mTOR signaling pathway negative regulator Tsc1 or Pten caused mouse pituitary prolactinoma, which was abolished by rapamycin treatment. Mechanistically, the expression of pituitary tumor transforming gene 1 (PTTG1) was upregulated in an mTOR complex 1-dependent manner. Overexpressed PTTG1 was crucial in hyperactive mTOR-mediated tumorigenesis. mTOR-PTTG1 signaling axis may be targeted for the treatment of tumors with mTOR hyperactivation.
Collapse
|
12
|
Zasadil LM, Britigan EMC, Ryan SD, Kaur C, Guckenberger DJ, Beebe DJ, Moser AR, Weaver BA. High rates of chromosome missegregation suppress tumor progression but do not inhibit tumor initiation. Mol Biol Cell 2016; 27:1981-9. [PMID: 27146113 PMCID: PMC4927272 DOI: 10.1091/mbc.e15-10-0747] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 04/23/2016] [Indexed: 11/11/2022] Open
Abstract
Aneuploidy, an abnormal chromosome number that deviates from a multiple of the haploid, has been recognized as a common feature of cancers for >100 yr. Previously, we showed that the rate of chromosome missegregation/chromosomal instability (CIN) determines the effect of aneuploidy on tumors; whereas low rates of CIN are weakly tumor promoting, higher rates of CIN cause cell death and tumor suppression. However, whether high CIN inhibits tumor initiation or suppresses the growth and progression of already initiated tumors remained unclear. We tested this using the Apc(Min/+) mouse intestinal tumor model, in which effects on tumor initiation versus progression can be discriminated. Apc(Min/+) cells exhibit low CIN, and we generated high CIN by reducing expression of the kinesin-like mitotic motor protein CENP-E. CENP-E(+/-);Apc(Min/+) doubly heterozygous cells had higher rates of chromosome missegregation than singly heterozygous cells, resulting in increased cell death and a substantial reduction in tumor progression compared with Apc(Min/+) animals. Intestinal organoid studies confirmed that high CIN does not inhibit tumor cell initiation but does inhibit subsequent cell growth. These findings support the conclusion that increasing the rate of chromosome missegregation could serve as a successful chemotherapeutic strategy.
Collapse
Affiliation(s)
- Lauren M Zasadil
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705 Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705
| | - Eric M C Britigan
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705 Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705
| | - Sean D Ryan
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705
| | - Charanjeet Kaur
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705
| | - David J Guckenberger
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705 Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705
| | - Amy R Moser
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705 Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705
| | - Beth A Weaver
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705 Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705
| |
Collapse
|
13
|
Lines KE, Stevenson M, Thakker RV. Animal models of pituitary neoplasia. Mol Cell Endocrinol 2016; 421:68-81. [PMID: 26320859 PMCID: PMC4721536 DOI: 10.1016/j.mce.2015.08.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 08/25/2015] [Accepted: 08/25/2015] [Indexed: 01/21/2023]
Abstract
Pituitary neoplasias can occur as part of a complex inherited disorder, or more commonly as sporadic (non-familial) disease. Studies of the molecular and genetic mechanisms causing such pituitary tumours have identified dysregulation of >35 genes, with many revealed by studies in mice, rats and zebrafish. Strategies used to generate these animal models have included gene knockout, gene knockin and transgenic over-expression, as well as chemical mutagenesis and drug induction. These animal models provide an important resource for investigation of tissue-specific tumourigenic mechanisms, and evaluations of novel therapies, illustrated by studies into multiple endocrine neoplasia type 1 (MEN1), a hereditary syndrome in which ∼ 30% of patients develop pituitary adenomas. This review describes animal models of pituitary neoplasia that have been generated, together with some recent advances in gene editing technologies, and an illustration of the use of the Men1 mouse as a pre clinical model for evaluating novel therapies.
Collapse
Affiliation(s)
- K E Lines
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - M Stevenson
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - R V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, Headington, Oxford OX3 7LJ, UK.
| |
Collapse
|
14
|
Molecular Mechanisms Underlying Pituitary Pathogenesis. Biochem Genet 2015; 54:107-19. [DOI: 10.1007/s10528-015-9709-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/19/2015] [Indexed: 10/22/2022]
|
15
|
Giam M, Rancati G. Aneuploidy and chromosomal instability in cancer: a jackpot to chaos. Cell Div 2015; 10:3. [PMID: 26015801 PMCID: PMC4443636 DOI: 10.1186/s13008-015-0009-7] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 05/08/2015] [Indexed: 12/12/2022] Open
Abstract
Genomic instability (GIN) is a hallmark of cancer cells that facilitates the acquisition of mutations conferring aggressive or drug-resistant phenotypes during cancer evolution. Chromosomal instability (CIN) is a form of GIN that involves frequent cytogenetic changes leading to changes in chromosome copy number (aneuploidy). While both CIN and aneuploidy are common characteristics of cancer cells, their roles in tumor initiation and progression are unclear. On the one hand, CIN and aneuploidy are known to provide genetic variation to allow cells to adapt in changing environments such as nutrient fluctuations and hypoxia. Patients with constitutive aneuploidies are more susceptible to certain types of cancers, suggesting that changes in chromosome copy number could positively contribute to cancer evolution. On the other hand, chromosomal imbalances have been observed to have detrimental effects on cellular fitness and might trigger cell cycle arrest or apoptosis. Furthermore, mouse models for CIN have led to conflicting results. Taken together these findings suggest that the relationship between CIN, aneuploidy and cancer is more complex than what was previously anticipated. Here we review what is known about this complex ménage à trois, discuss recent evidence suggesting that aneuploidy, CIN and GIN together promote a vicious cycle of genome chaos. Lastly, we propose a working hypothesis to reconcile the conflicting observations regarding the role of aneuploidy and CIN in tumorigenesis.
Collapse
Affiliation(s)
- Maybelline Giam
- Institute for Medical Biology (IMB), Agency for Science, Technology and Research (ASTAR), Singapore, 138648 Singapore
| | - Giulia Rancati
- Institute for Medical Biology (IMB), Agency for Science, Technology and Research (ASTAR), Singapore, 138648 Singapore ; School of Biological Sciences, Nanyang Technological University, Singapore, 637551 Singapore ; Department of Biochemistry, Yong Loo Lin School of Medicine, NUS, Singapore, 117597 Singapore
| |
Collapse
|
16
|
Xekouki P, Szarek E, Bullova P, Giubellino A, Quezado M, Mastroyannis SA, Mastorakos P, Wassif CA, Raygada M, Rentia N, Dye L, Cougnoux A, Koziol D, Sierra MDLL, Lyssikatos C, Belyavskaya E, Malchoff C, Moline J, Eng C, Maher LJ, Pacak K, Lodish M, Stratakis CA. Pituitary adenoma with paraganglioma/pheochromocytoma (3PAs) and succinate dehydrogenase defects in humans and mice. J Clin Endocrinol Metab 2015; 100:E710-9. [PMID: 25695889 PMCID: PMC4422891 DOI: 10.1210/jc.2014-4297] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
CONTEXT Germline mutations in genes coding succinate dehydrogenase (SDH) subunits A, B, C, and D have been identified in familial paragangliomas (PGLs)/pheochromocytomas (PHEOs) and other tumors. We described a GH-secreting pituitary adenoma (PA) caused by SDHD mutation in a patient with familial PGLs. Additional patients with PAs and SDHx defects have since been reported. DESIGN We studied 168 patients with unselected sporadic PA and with the association of PAs, PGLs, and/or pheochromocytomas, a condition we named the 3P association (3PAs) for SDHx germline mutations. We also studied the pituitary gland and hormonal profile of Sdhb(+/-) mice and their wild-type littermates at different ages. RESULTS No SDHx mutations were detected among sporadic PA, whereas three of four familial cases were positive for a mutation (75%). Most of the SDHx-deficient PAs were either prolactinomas or somatotropinomas. Pituitaries of Sdhb(+/-) mice older than 12 months had an increased number mainly of prolactin-secreting cells and several ultrastructural abnormalities such as intranuclear inclusions, altered chromatin nuclear pattern, and abnormal mitochondria. Igf-1 levels of mutant mice tended to be higher across age groups, whereas Prl and Gh levels varied according to age and sex. CONCLUSION The present study confirms the existence of a new association that we termed 3PAs. It is due mostly to germline SDHx defects, although sporadic cases of 3PAs without SDHx defects also exist. Using Sdhb(+/-) mice, we provide evidence that pituitary hyperplasia in SDHx-deficient cells may be the initial abnormality in the cascade of events leading to PA formation.
Collapse
Affiliation(s)
- Paraskevi Xekouki
- Section on Endocrinology and Genetics (P.X., E.S., S.A.M., P.M., M.R., N.R., M.d.L.L.S., C.L., E.B., M.L., C.A.S.), Program on Developmental Endocrinology and Genetics, Section on Medical Neuroendocrinology (P.B., A.G.), Program in Reproductive and Adult Endocrinology, Section on Molecular Dysmorphology (C.A.W., A.C.), Program in Developmental Endocrinology and Genetics, Microscopy and Imaging Core (L.D.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Laboratory of Pathology (A.G., M.Q., K.P.), National Cancer Institute, and Biostatistics and Clinical Epidemiology Service (D.K.), Clinical Center, National Institutes of Health, Bethesda, Maryland 20892; Department of Endocrinology (C.M.), University of Connecticut Health Center, Farmington, Connecticut 06030; Genomic Medicine Institute (J.M., C.E.), Cleveland Clinic, Cleveland, Ohio 44195; Department of Biochemistry and Molecular Biology (L.J.M.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905; and Department of Molecular Medicine (P.B.), Institute of Virology, Slovak Academy of Sciences, 833 06 Bratislava, Slovakia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Fukuoka H. New potential targets for treatment of Cushing's disease: epithelial growth factor receptor and cyclin-dependent kinases. Pituitary 2015; 18:274-8. [PMID: 25612787 DOI: 10.1007/s11102-015-0637-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Cushing's disease (CD) is caused by adrenocorticotropic hormone (ACTH)-producing pituitary adenomas (ACTHomas). Drug treatment for CD consists of three strategies: pituitary tumor-targeted therapy, steroidogenesis inhibitors, and glucocorticoid receptor antagonists. All of these strategies are under development, and several new drugs have recently been approved for clinical use or are being tested in clinical trials. Pituitary-targeted drugs are a particularly important method in the treatment of CD. Available pituitary tumor-targeted drugs include a dopamine receptor agonist and a somatostatin analog. Since disrupted cell cycle signaling is clearly associated with pathogenesis of ACTHomas which express active forms of epithelial growth factor receptor (EGFR), cyclins, and the catalytic subunit of cyclin-dependent kinases (CDKs), we focused on these molecules as therapeutic targets for ACTHomas. METHODS In this review, a literature search were performed using PubMed with following terms; Cushing's disease, EGFR, CDKs, cell cycle, and targeted therapy. CONCLUSION Accumulating evidence demonstrates that EGFR and cyclin E-CDK2 may be promising targets for treating ACTHomas.
Collapse
Affiliation(s)
- Hidenori Fukuoka
- Division of Diabetes and Endocrinology, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan,
| |
Collapse
|
18
|
Abstract
Chromosomal instability (CIN) is a process leading to errors in chromosome segregation and results in aneuploidy, a state in which cells have an abnormal number of chromosomes. CIN is a hallmark of cancer, and furthermore linked to ageing and age-related diseases such as Alzheimer's. Various mouse models have been developed to explore the role of CIN in ageing and cancer. While these models reveal only a modest contribution of CIN to the initiation of cancer, they also clearly show that CIN is a powerful accelerator of cancer in a predisposed background. Other than cancer, CIN also appears to provoke premature ageing in some of the CIN models. In this review, we discuss the phenotypes of the various available mouse models, what we have learnt so far, and importantly, also which questions still need to be addressed.
Collapse
|
19
|
Takekoshi S, Yasui Y, Inomoto C, Kitatani K, Nakamura N, Osamura RY. A Histopathological Study of Multi-hormone Producing Proliferative Lesions in Estrogen-induced Rat Pituitary Prolactinoma. Acta Histochem Cytochem 2014; 47:155-64. [PMID: 25392569 PMCID: PMC4164703 DOI: 10.1267/ahc.14029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 06/10/2014] [Indexed: 01/23/2023] Open
Abstract
Rats with estrogen-induced prolactin-producing pituitary adenoma (E2-PRLoma) have been employed as an animal model of human PRL-producing pituitary adenoma in a large number of studies. Presently, we found that long-term administration of estrogen to SD rats resulted in the development of E2-PRLomas, some of which included multi-hormone producing nodules. We herein report results of histopathological analyses of these lesions. PRLoma models were created in female SD rats by 22 weeks or longer administration of a controlled-release preparation of estradiol at a dose of 10 mg/kg/2 weeks. Ten of the 11 PRLoma model rats had proliferative nodular lesions composed of large eosinophilic cells like gonadotrophs inside the PRLoma. These lesions were positive for PRL, TSHβ, and α subunits and were negative for GH, LHβ, ACTH, and S-100. Double immunostaining revealed that these large eosinophilic cells showed coexpression of PRL and TSHβ, PRL and α subunits, and TSHβ and α subunits. Those results clarified that long-term estrogen administration to female SD rats induced multi-hormone producing neoplastic pituitary nodules that expressed PRL, TSHβ, and α subunits. We studied these neoplastic nodules obtained by laser microdissection to acquire findings similar to those of the immunohistochemical analysis. We consider that this animal model is useful for pathogenesis analyses and therapeutic agent development concerning human multi-hormone producing pituitary adenomas.
Collapse
Affiliation(s)
- Susumu Takekoshi
- Department of Cell Biology, Division of Host Defense Mechanism, Tokai University School of Medicine
| | - Yuzo Yasui
- Department of Cell Biology, Division of Host Defense Mechanism, Tokai University School of Medicine
| | - Chie Inomoto
- Department of Pathology, Tokai University School of Medicine
| | - Kanae Kitatani
- Department of Cell Biology, Division of Host Defense Mechanism, Tokai University School of Medicine
| | - Naoya Nakamura
- Department of Pathology, Tokai University School of Medicine
| | | |
Collapse
|
20
|
Cano DA, Soto-Moreno A, Leal-Cerro A. Genetically engineered mouse models of pituitary tumors. Front Oncol 2014; 4:203. [PMID: 25136513 PMCID: PMC4117927 DOI: 10.3389/fonc.2014.00203] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 07/15/2014] [Indexed: 12/28/2022] Open
Abstract
Animal models constitute valuable tools for investigating the pathogenesis of cancer as well as for preclinical testing of novel therapeutics approaches. However, the pathogenic mechanisms of pituitary-tumor formation remain poorly understood, particularly in sporadic adenomas, thus, making it a challenge to model pituitary tumors in mice. Nevertheless, genetically engineered mouse models (GEMMs) of pituitary tumors have provided important insight into pituitary tumor biology. In this paper, we review various GEMMs of pituitary tumors, highlighting their contributions and limitations, and discuss opportunities for research in the field.
Collapse
Affiliation(s)
- David A Cano
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen del Rocío , Seville , Spain ; Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla , Seville , Spain
| | - Alfonso Soto-Moreno
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen del Rocío , Seville , Spain ; Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla , Seville , Spain
| | - Alfonso Leal-Cerro
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla , Seville , Spain
| |
Collapse
|
21
|
Li Y, Zhou LP, Ma P, Sui CG, Meng FD, Tian X, Fu LY, Jiang YH. Relationship of PTTG Expression with Tumor Invasiveness and Microvessel Density of Pituitary Adenomas: A Meta-Analysis. Genet Test Mol Biomarkers 2014; 18:279-85. [PMID: 24611443 DOI: 10.1089/gtmb.2013.0447] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Yan Li
- Molecular Oncology Department of Cancer Research Institution, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Li-Ping Zhou
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Ping Ma
- Molecular Oncology Department of Cancer Research Institution, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Cheng-Guang Sui
- Molecular Oncology Department of Cancer Research Institution, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Fan-Dong Meng
- Molecular Oncology Department of Cancer Research Institution, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xin Tian
- Molecular Oncology Department of Cancer Research Institution, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Li-Ye Fu
- Molecular Oncology Department of Cancer Research Institution, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - You-Hong Jiang
- Molecular Oncology Department of Cancer Research Institution, The First Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
22
|
Donangelo I, Ren SG, Eigler T, Svendsen C, Melmed S. Sca1⁺ murine pituitary adenoma cells show tumor-growth advantage. Endocr Relat Cancer 2014; 21:203-16. [PMID: 24481638 PMCID: PMC3978815 DOI: 10.1530/erc-13-0229] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The role of tumor stem cells in benign tumors such as pituitary adenomas remains unclear. In this study, we investigated whether the cells within pituitary adenomas that spontaneously develop in Rb+/- mice are hierarchically distributed with a subset being responsible for tumor growth. Cells derived directly from such tumors grew as spheres in serum-free culture medium supplemented with epidermal growth factor and basic fibroblast growth factor. Some cells within growing pituitary tumor spheres (PTS) expressed common stem cell markers (Sca1, Sox2, Nestin, and CD133), but were devoid of hormone-positive differentiated cells. Under subsequent differentiating conditions (matrigel-coated growth surface), PTS expressed all six pituitary hormones. We next searched for specific markers of the stem cell population and isolated a Sca1(+) cell population that showed increased sphere formation potential, lower mRNA hormone expression, higher expression of stem cell markers (Notch1, Sox2, and Nestin), and increased proliferation rates. When transplanted into non-obese diabetic-severe combined immunodeficiency gamma mice brains, Sca1(+) pituitary tumor cells exhibited higher rates of tumor formation (brain tumors observed in 11/11 (100%) vs 7/12 (54%) of mice transplanted with Sca1(+) and Sca1(-) cells respectively). Magnetic resonance imaging and histological analysis of brain tumors showed that tumors derived from Sca1(+) pituitary tumor cells were also larger and plurihormonal. Our findings show that Sca1(+) cells derived from benign pituitary tumors exhibit an undifferentiated expression profile and tumor-proliferative advantages, and we propose that they could represent putative pituitary tumor stem/progenitor cells.
Collapse
Affiliation(s)
| | | | | | - Clive Svendsen
- Regenerative Medicine Institute, Cedars-Sinai Medical Center
| | | |
Collapse
|
23
|
Correlations of pituitary tumor transforming gene expression with human pituitary adenomas: a meta-analysis. PLoS One 2014; 9:e90396. [PMID: 24594688 PMCID: PMC3942425 DOI: 10.1371/journal.pone.0090396] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 01/29/2014] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE Pituitary tumor transforming gene (PTTG) is an important paracrine growth factor involved in early lactotrope transformation and early onset of angiogenesis in pituitary hyperplasia. Emerging evidences have shown that PTTG expression may contribute to the etiology of pituitary adenomas; but individually published studies showed inconclusive results. This meta-analysis aimed to derive a more precise estimation of the correlations of PTTG expression with human pituitary adenomas. METHODS A range of electronic databases were searched: MEDLINE (1966∼2013), the Cochrane Library Database (Issue 12, 2013), EMBASE (1980∼2013), CINAHL (1982∼2013), Web of Science (1945∼2013) and the Chinese Biomedical Database (CBM) (1982∼2013) without language restrictions. Meta-analysis was performed using the STATA 12.0 software. Crude odds ratio (OR) or standard mean difference (SMD) with its corresponding 95% confidence interval (95%CI) were calculated. RESULTS Twenty-four clinical cohort studies were included with a total of 1,464 pituitary adenomas patients. The meta-analysis results revealed that patients with invasive pituitary adenomas had higher positive expression of PTTG than those of non-invasive patients (OR = 6.68, 95%CI = 3.72-11.99, P<0.001). We also found a significant difference in microvessel density between invasive and non-invasive patients (SMD = 1.81, 95%CI = 0.39-3.23, P = 0.013). However, there were no significant difference in PTTG expression between functional and non-functional patients with pituitary adenomas (OR = 1.11, 95%CI = 0.58-2.10, P = 0.753). No publication bias was detected in this meta-analysis (all P>0.05). CONCLUSION This present meta-analysis suggests that PTTG expression may be associated with tumor invasiveness and microvessel density of pituitary adenomas, while no correlations with functional status was found.
Collapse
|
24
|
Growth hormone is a cellular senescence target in pituitary and nonpituitary cells. Proc Natl Acad Sci U S A 2013; 110:E3331-9. [PMID: 23940366 DOI: 10.1073/pnas.1310589110] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Premature proliferative arrest in benign or early-stage tumors induced by oncoproteins, chromosomal instability, or DNA damage is associated with p53/p21 activation, culminating in either senescence or apoptosis, depending on cell context. Growth hormone (GH) elicits direct peripheral metabolic actions as well as growth effects mediated by insulin-like growth factor 1 (IGF1). Locally produced peripheral tissue GH, in contrast to circulating pituitary-derived endocrine GH, has been proposed to be both proapoptotic and prooncogenic. Pituitary adenomas expressing and secreting GH are invariably benign and exhibit DNA damage and a senescent phenotype. We therefore tested effects of nutlin-induced p53-mediated senescence in rat and human pituitary cells. We show that DNA damage senescence induced by nutlin triggers the p53/p21 senescent pathway, with subsequent marked induction of intracellular pituitary GH in vitro. In contrast, GH is not induced in cells devoid of p53. Furthermore we show that p53 binds specific GH promoter motifs and enhances GH transcription and secretion in senescent pituitary adenoma cells and also in nonpituitary (human breast and colon) cells. In vivo, treatment with nutlin results in up-regulation of both p53 and GH in the pituitary gland, as well as increased GH expression in nonpituitary tissues (lung and liver). Intracrine GH acts in pituitary cells as an apoptosis switch for p53-mediated senescence, likely protecting the pituitary adenoma from progression to malignancy. Unlike in the pituitary, in nonpituitary cells GH exerts antiapoptotic properties. Thus, the results show that GH is a direct p53 transcriptional target and fulfills criteria as a p53 target gene. Induced GH is a readily measurable cell marker for p53-mediated cellular senescence.
Collapse
|
25
|
Fong MY, Farghaly H, Kakar SS. Tumorigenic potential of pituitary tumor transforming gene (PTTG) in vivo investigated using a transgenic mouse model, and effects of cross breeding with p53 (+/-) transgenic mice. BMC Cancer 2012; 12:532. [PMID: 23164239 PMCID: PMC3530432 DOI: 10.1186/1471-2407-12-532] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 11/08/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pituitary tumor-transforming gene (PTTG) is an oncogene that is overexpressed in variety of tumors and exhibits characteristics of a transforming gene. Previous transgenic mouse models to access the tumorigenic potential in the pituitary and ovary have resulted in dysplasia without formation of visible tumors, possibly due to the insufficient expression of PTTG. PTTG expression level is critical for ovarian tumorigenesis in a xenograft model. Therefore, the tumorigenic function of PTTG in vivo remains unclear. We generated a transgenic mouse that overexpresses PTTG driven by the CMV promoter to determine whether PTTG functions as a transforming oncogene that is capable of initiating tumorigenesis. METHODS Transgenic animals were generated by microinjection of PTTG transgene into the male pronucleus of FVB 0.5 day old embryos. Expression levels of PTTG in tissues of transgenic animals were analyzed using an immunohistochemical analysis. H&E staining and immunohistostaining were performed to examine the type of tumor in transgenic and PTTG transgenic/p53+/- animals. RESULTS PTTG transgenic offspring (TgPTTG) were monitored for tumor development at various ages. H&E analysis was performed to identify the presence of cancer and hyperplastic conditions verified with the proliferation marker PCNA and the microvessel marker CD31. Immunohistochemistry was performed to determine transgene expression, revealing localization to the epithelium of the fallopian tube, with more generalized expression in the liver, lung, kidney, and spleen. At eight months of age, 2 out of 15 TgPTTG developed ovarian cancer, 2 out of 15 developed benign tumors, 2 out of 15 developed cervical dysplasia, and 3 out of 15 developed adenomyosis of the uterus. At ten months of age, 2 out of 10 TgPTTG developed adenocarcinoma of the ovary, 1 out of 10 developed a papillary serous adenocarcinoma, and 2 out of 10 presented with atypia of ovarian epithelial cells. Tumorigenesis is a multi-step process, often requiring multiple oncogenes and/or inactivation of tumor suppressor genes. Therefore, to understand the contribution of p53 to PTTG induced tumorigenesis, we crossbred TgPTTG to p53+/- mice and maintained those 8 to 10 months. TgPTTG/p53+/- animals developed sarcomas faster than p53+/- alone as well as different tumor types in addition to cervical carcinomas in situ in 10 out of 17 females. CONCLUSIONS We conclude that while PTTG is a functional transforming oncogene, it requires an additional partner to effectively promote tumorigenesis through the loss of p53 include or between function or modulation.
Collapse
Affiliation(s)
- Miranda Y Fong
- Department of Physiology and Biophysics, University of Louisville, 505 South Hancock Street, CTRB 322, Louisville, KY 40202, USA
| | | | | |
Collapse
|
26
|
Chesnokova V, Zonis S, Wawrowsky K, Tani Y, Ben-Shlomo A, Ljubimov V, Mamelak A, Bannykh S, Melmed S. Clusterin and FOXL2 act concordantly to regulate pituitary gonadotroph adenoma growth. Mol Endocrinol 2012; 26:2092-103. [PMID: 23051594 DOI: 10.1210/me.2012-1158] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Pituitary tumors grow slowly and despite their high prevalence are invariably benign. We therefore studied mechanisms underlying pituitary tumor growth restraint. Pituitary tumor transforming gene (PTTG), the index human securin, a hallmark of pituitary tumors, triggers pituitary cell proliferation and murine pituitary tumor development. We show that human gonadotroph cell pituitary tumors, unlike other secreting tumor types, express high levels of gonadotroph-specific forkhead transcription factor FOXL2, and both PTTG and Forkhead box protein L2 (FOXL2) stimulate gonadotroph clusterin (Clu) expression. Both Clu RNA isoforms are abundantly expressed in these nonhormone-secreting human tumors, and, when cultured, these tumor cells release highly abundant levels of secreted Clu. FOXL2 directly stimulates the Clu gene promoter, and we show that PTTG triggers ataxia telangiectasia mutated kinase/IGF-I/p38MAPK DNA damage/chromosomal instability signaling, which in turn also induces Clu expression. Consequently, Clu restrains pituitary cell proliferation by inducing cyclin dependent kinase inhibitors p16 and p27, whereas Clu deletion down-regulates p16 and p27 in the Clu(-/-) mouse pituitary. FOXL2 binds and suppresses the PTTG promoter, and Clu also suppresses PTTG expression, thus neutralizing protumorigenic PTTG gonadotroph tumor cell properties. In vivo, murine gonadotroph LβT2 tumor cell xenografts overexpressing Clu and FOXL2 both grow slower and elicit smaller tumors. Thus, gonadotroph tumor cell proliferation is determined by the interplay between cell-specific FOXL2 with PTTG and Clu.
Collapse
Affiliation(s)
- Vera Chesnokova
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Tong Y, Zhao W, Zhou C, Wawrowsky K, Melmed S. PTTG1 attenuates drug-induced cellular senescence. PLoS One 2011; 6:e23754. [PMID: 21858218 PMCID: PMC3157437 DOI: 10.1371/journal.pone.0023754] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 07/24/2011] [Indexed: 12/15/2022] Open
Abstract
As PTTG1 (pituitary tumor transforming gene) abundance correlates with adverse outcomes in cancer treatment, we determined mechanisms underlying this observation by assessing the role of PTTG1 in regulating cell response to anti-neoplastic drugs. HCT116 cells devoid of PTTG1 (PTTG1−/−) exhibited enhanced drug sensitivity as assessed by measuring BrdU incorporation in vitro. Apoptosis, mitosis catastrophe or DNA damage were not detected, but features of senescence were observed using low doses of doxorubicin and TSA. The number of drug-induced PTTG1−/− senescent cells increased ∼4 fold as compared to WT PTTG1-replete cells (p<0.001). p21, an important regulator of cell senescence, was induced ∼3 fold in HCT116 PTTG1−/− cells upon doxorubicin or Trichostatin A treatment. Binding of Sp1, p53 and p300 to the p21 promoter was enhanced in PTTG1−/− cells after treatment, suggesting transcriptional regulation of p21. p21 knock down abrogated the observed senescent effects of these drugs, indicating that PTTG1 likely suppresses p21 to regulate drug-induced senescence. PTTG1 also regulated SW620 colon cancer cells response to doxorubicin and TSA mediated by p21. Subcutaneously xenografted PTTG1−/− HCT116 cells developed smaller tumors and exhibited enhanced responses to doxorubicin. PTTG1−/− tumor tissue derived from excised tumors exhibited increased doxorubicin-induced senescence. As senescence is a determinant of cell responses to anti-neoplastic treatments, these findings suggest PTTG1 as a tumor cell marker to predict anti-neoplastic treatment outcomes.
Collapse
Affiliation(s)
- Yunguang Tong
- Department of Medicine, Cedars-Sinai Medical Center, University of California Los Angeles, Los Angeles, California, United States of America
| | - Weijiang Zhao
- Department of Medicine, Cedars-Sinai Medical Center, University of California Los Angeles, Los Angeles, California, United States of America
| | - Cuiqi Zhou
- Department of Medicine, Cedars-Sinai Medical Center, University of California Los Angeles, Los Angeles, California, United States of America
| | - Kolja Wawrowsky
- Department of Medicine, Cedars-Sinai Medical Center, University of California Los Angeles, Los Angeles, California, United States of America
| | - Shlomo Melmed
- Department of Medicine, Cedars-Sinai Medical Center, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
28
|
Targeting zebrafish and murine pituitary corticotroph tumors with a cyclin-dependent kinase (CDK) inhibitor. Proc Natl Acad Sci U S A 2011; 108:8414-9. [PMID: 21536883 DOI: 10.1073/pnas.1018091108] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cushing disease caused by adrenocorticotropin (ACTH)-secreting pituitary adenomas leads to hypercortisolemia predisposing to diabetes, hypertension, osteoporosis, central obesity, cardiovascular morbidity, and increased mortality. There is no effective pituitary targeted pharmacotherapy for Cushing disease. Here, we generated germline transgenic zebrafish with overexpression of pituitary tumor transforming gene (PTTG/securin) targeted to the adenohypophyseal proopiomelanocortin (POMC) lineage, which recapitulated early features pathognomonic of corticotroph adenomas, including corticotroph expansion and partial glucocorticoid resistance. Adult Tg:Pomc-Pttg fish develop neoplastic coticotrophs and pituitary cyclin E up-regulation, as well as metabolic disturbances mimicking hypercortisolism caused by Cushing disease. Early development of corticotroph pathologies in Tg:Pomc-Pttg embryos facilitated drug testing in vivo. We identified a pharmacologic CDK2/cyclin E inhibitor, R-roscovitine (seliciclib; CYC202), which specifically reversed corticotroph expansion in live Tg:Pomc-Pttg embryos. We further validated that orally administered R-roscovitine suppresses ACTH and corticosterone levels, and also restrained tumor growth in a mouse model of ACTH-secreting pituitary adenomas. Molecular analyses in vitro and in vivo showed that R-roscovitine suppresses ACTH expression, induces corticotroph tumor cell senescence and cell cycle exit by up-regulating p27, p21 and p57, and downregulates cyclin E expression. The results suggest that use of selective CDK inhibitors could effectively target corticotroph tumor growth and hormone secretion.
Collapse
|
29
|
Chen S, Xiao L, Liu Z, Liu J, Liu Y. Pituitary tumor transforming gene-1 haplotypes and risk of pituitary adenoma: a case-control study. BMC MEDICAL GENETICS 2011; 12:44. [PMID: 21439054 PMCID: PMC3078851 DOI: 10.1186/1471-2350-12-44] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 03/25/2011] [Indexed: 11/10/2022]
Abstract
Background It has been suggested that pituitary adenoma results from accumulation of multiple genetic and/or epigenetic aberrations, which may be identified through association studies. As pituitary tumor transforming gene-1 (PTTG1)/securin plays a critical role in promoting genomic instability in pituitary neoplasia, the present study explored the association of PTTG1 haplotypes with the risk of pituitary adenoma. Methods We genotyped five PTTG1 haplotype-tagging SNPs (htSNP) by PCR-RFLP assays in a case-control study, which included 280 Han Chinese patients diagnosed with pituitary adenoma and 280 age-, gender- and geographically matched Han Chinese controls. Haplotypes were reconstructed according to the genotyping data and linkage disequilibrium status of the htSNPs. Results No significant differences in allele and genotype frequencies of the htSNPs were observed between pituitary adenoma patients and controls, indicating that none of the individual PTTG1 SNPs examined in this study is associated with the risk of pituitary adenoma. In addition, no significant association was detected between the reconstructed PTTG1 haplotypes and pituitary adenoma cases or the controls. Conclusions Though no significant association was found between PTTG1 haplotypes and the risk of pituitary adenoma, this is the first report on the association of individual PTTG1 SNPs or PTTG1 haplotypes with the risk of pituitary adenoma based on a solid study; it will provide an important reference for future studies on the association between genetic alterations in PTTG1 and the risk of pituitary adenoma or other tumors.
Collapse
Affiliation(s)
- Shuai Chen
- Gamma Knife Treatment and Research Center, Xiangya Hospital, Central South University, Changsha, 410008 PR China
| | | | | | | | | |
Collapse
|
30
|
Chesnokova V, Zonis S, Zhou C, Ben-Shlomo A, Wawrowsky K, Toledano Y, Tong Y, Kovacs K, Scheithauer B, Melmed S. Lineage-specific restraint of pituitary gonadotroph cell adenoma growth. PLoS One 2011; 6:e17924. [PMID: 21464964 PMCID: PMC3064664 DOI: 10.1371/journal.pone.0017924] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 02/15/2011] [Indexed: 11/23/2022] Open
Abstract
Although pituitary adenomas are usually benign, unique trophic mechanisms restraining cell proliferation are unclear. As GH-secreting adenomas are associated with p53/p21-dependent senescence, we tested mechanisms constraining non-functioning pituitary adenoma growth. Thirty six gonadotroph-derived non-functioning pituitary adenomas all exhibited DNA damage, but undetectable p21 expression. However, these adenomas all expressed p16, and >90% abundantly expressed cytoplasmic clusterin associated with induction of the Cdk inhibitor p15 in 70% of gonadotroph and in 26% of somatotroph lineage adenomas (p = 0.006). Murine LβT2 and αT3 gonadotroph pituitary cells, and αGSU.PTTG transgenic mice with targeted gonadotroph cell adenomas also abundantly expressed clusterin and exhibited features of oncogene-induced senescence as evidenced by C/EBPβ and C/EBPδ induction. In turn, C/EBPs activated the clusterin promoter ∼5 fold, and elevated clusterin subsequently elicited p15 and p16 expression, acting to arrest murine gonadotroph cell proliferation. In contrast, specific clusterin suppression by RNAis enhanced gonadotroph proliferation. FOXL2, a tissue-specific gonadotroph lineage factor, also induced the clusterin promoter ∼3 fold in αT3 pituitary cells. As nine of 12 pituitary carcinomas were devoid of clusterin expression, this protein may limit proliferation of benign adenomatous pituitary cells. These results point to lineage-specific pathways restricting uncontrolled murine and human pituitary gonadotroph adenoma cell growth.
Collapse
Affiliation(s)
- Vera Chesnokova
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Svetlana Zonis
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Cuiqi Zhou
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Anat Ben-Shlomo
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Kolja Wawrowsky
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Yoel Toledano
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Yunguang Tong
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Departments of Pathology, St. Michael's Hospital, Toronto, Canada
- Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kalman Kovacs
- Departments of Pathology, St. Michael's Hospital, Toronto, Canada
| | | | - Shlomo Melmed
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
31
|
Bacac M, Fusco C, Planche A, Santodomingo J, Demaurex N, Leemann-Zakaryan R, Provero P, Stamenkovic I. Securin and separase modulate membrane traffic by affecting endosomal acidification. Traffic 2011; 12:615-26. [PMID: 21272169 DOI: 10.1111/j.1600-0854.2011.01169.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Securin and separase play a key role in sister chromatid separation during anaphase. However, a growing body of evidence suggests that in addition to regulating chromosome segregation, securin and separase display functions implicated in membrane traffic in Caenorhabditis elegans and Drosophila. Here we show that in mammalian cells both securin and separase associate with membranes and that depletion of either protein causes robust swelling of the trans-Golgi network (TGN) along with the appearance of large endocytic vesicles in the perinuclear region. These changes are accompanied by diminished constitutive protein secretion as well as impaired receptor recycling and degradation. Unexpectedly, cells depleted of securin or separase display defective acidification of early endosomes and increased membrane recruitment of vacuolar (V-) ATPase complexes, mimicking the effect of the specific V-ATPase inhibitor Bafilomycin A1. Taken together, our findings identify a new functional role of securin and separase in the modulation of membrane traffic and protein secretion that implicates regulation of V-ATPase assembly and function.
Collapse
Affiliation(s)
- Marina Bacac
- Division of Experimental Pathology, Faculty of Biology and Medicine, Institute of Pathology, CHUV, University of Lausanne, Rue du Bugnon 25, Lausanne CH1011, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
The pituitary tumor-transforming gene (PTTG1) encodes a multifunctional protein (PTTG) that is overexpressed in numerous tumours, including pituitary, thyroid, breast and ovarian carcinomas. PTTG induces cellular transformation in vitro and tumourigenesis in vivo, and several mechanisms by which PTTG contributes to tumourigenesis have been investigated. Also known as the human securin, PTTG is involved in cell cycle regulation, controlling the segregation of sister chromatids during mitosis. This review outlines current information regarding PTTG structure, expression, regulation and function in the pathogenesis of neoplasia. Recent progress concerning the use of PTTG as a prognostic marker or therapeutic target will be considered. In addition, the PTTG binding factor (PBF), identified through its interaction with PTTG, has also been established as a proto-oncogene that is upregulated in several cancers. Current knowledge regarding PBF is outlined and its role both independently and alongside PTTG in endocrine and related cancers is discussed.
Collapse
|
33
|
Abstract
The majority of patients with acromegaly harbor a functioning growth hormone (GH) pituitary adenoma. Growth hormone–secreting adenomas correspond to about 20% of all pituitary adenomas. From the histopathological point of view, a variety of adenomas may present with clinical signs and symptoms of GH hypersecretion including pure GH cell adenomas (densely and sparsely granulated GH adenomas), mixed GH and prolactin cell adenomas, and monomorphous adenomas with primitive cells able to secrete GH and prolactin including the acidophilic stem cell adenoma and the mammosomatotroph cell adenoma. In this article, the author reviews the main pathological features of the GH-secreting adenomas and some of the molecular genetics mechanisms involved in their pathogenesis.
Collapse
|
34
|
Abstract
Since the onset of the genomic era, there has been tremendous progress in identifying the genetic causes of endocrine tumours. Although this knowledge is valuable in its own right, understanding the molecular basis of tumourigenesis allows the development of new therapies targeted at the causative defects. Understanding the connection between genotype and phenotype is a complex process, which can only be partially understood from the analysis of primary tumours or from the studies of cells in vitro. To bridge this gap, genetically modified mice have been developed to allow molecular dissection of the relevant defects in an intact organism. In this article, we discuss the status of genetic modelling for hereditary and sporadic endocrine tumourigenesis with a goal towards providing a view of how this technology will be of future benefit to clinicians developing specifically targeted therapies for endocrine tumours.
Collapse
|
35
|
Salehi F, Kovacs K, Scheithauer BW, Cantelmi D, Horvath E, Lloyd RV, Cusimano M. Immunohistochemical expression of pituitary tumor transforming gene (PTTG) in pituitary adenomas: a correlative study of tumor subtypes. Int J Surg Pathol 2010; 18:5-13. [PMID: 20106827 DOI: 10.1177/1066896909356105] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE We investigated the correlation between immunohistochemical expression of the pituitary tumor transforming gene (PTTG) and pituitary adenoma subtype. METHODS Pituitary adenomas (n = 89) were stained for PTTG using the streptavidin-biotin-peroxidase complex method and a monoclonal PTTG antibody. RESULTS PTTG staining was found to be cytoplasmic with a pronounced paranuclear expression pattern. Reactivity was highest in growth hormone (GH) adenomas as compared with other tumors, including prolactin (PRL), follicle-stimulating hormone/luteinizing hormone/alpha subunit, as well as adrenocorticotrophic hormone-secreting adenomas. PRL adenomas exhibited the lowest expression levels. Among GH adenomas, untreated tumors demonstrated significantly higher PTTG levels than octreotide-treated examples. Although dopamine agonist-treated PRL adenomas tended to show lower expression levels, statistical significance was not reached. CONCLUSIONS Our finding that PTTG was differentially expressed in pituitary adenoma subtypes suggests a cell-specific function for PTTG. Moreover, treatment of GH adenomas with somatostatin analogues lowered PTTG expression. Further investigation into mechanisms mediating cell-specific expression of PTTG is warranted.
Collapse
Affiliation(s)
- Fateme Salehi
- St Michael's Hospital, University of Toronto, Toronto, ON, Canada.
| | | | | | | | | | | | | |
Collapse
|
36
|
Schvartzman JM, Sotillo R, Benezra R. Mitotic chromosomal instability and cancer: mouse modelling of the human disease. Nat Rev Cancer 2010; 10:102-15. [PMID: 20094045 PMCID: PMC5526619 DOI: 10.1038/nrc2781] [Citation(s) in RCA: 334] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The stepwise progression from an early dysplastic lesion to full-blown metastatic malignancy is associated with increases in genomic instability. Mitotic chromosomal instability - the inability to faithfully segregate equal chromosome complements to two daughter cells during mitosis - is a widespread phenomenon in solid tumours that is thought to serve as the fuel for tumorigenic progression. How chromosome instability (CIN) arises in tumours and what consequences it has are still, however, hotly debated issues. Here we review the recent literature with an emphasis on models that recapitulate observations from human disease.
Collapse
Affiliation(s)
- Juan-Manuel Schvartzman
- Program in Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | | | | |
Collapse
|
37
|
Abstract
In pituitary tumorigenesis there is cross-talk between fine deregulation of intracellular pathways and complex microenvironmental factors, processes that can be modulated at various levels. The signaling pathways of growth, angiogenic factors and hormones are intricate; therefore, alterations induced upon node-molecules can lead to aberrant proliferation. The demonstrated overactivity of AKT and MAPK pathways qualifies them as valuable targets for inhibition mediated by somatostatin analogues. An increasing body of evidence suggests clinically significant implications of PTTG1 in correlation with aggressive phenotypes or survival rate, thus PTTG1 is an interesting candidate biomarker for malignancy, tumor staging and subsequent therapeutic interventions. Future work should focus on understanding the molecular mechanisms that control pituitary tumor transformation, where intracellular signaling molecules will constitute not only diagnostic/prognostic markers but also novel therapeutic targets.
Collapse
Affiliation(s)
- Cristiana Pistol Tanase
- Victor Babes National Institute of Pathology, 99-101 Splaiul Independentei, Sector 5, Bucharest, Romania.
| | | | | |
Collapse
|
38
|
Zhou C, Wawrowsky K, Bannykh S, Gutman S, Melmed S. E2F1 induces pituitary tumor transforming gene (PTTG1) expression in human pituitary tumors. Mol Endocrinol 2009; 23:2000-12. [PMID: 19837943 DOI: 10.1210/me.2009-0161] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Rb/E2F is dysregulated in murine and human pituitary tumors. Pituitary tumor transforming gene (PTTG1), a securin protein, is required for pituitary tumorigenesis, and PTTG1 deletion attenuates pituitary tumor development in Rb(+/-) mice. E2F1 and PTTG1 were concordantly overexpressed in 29 of 46 Rb(+/-) murine pituitary tissues and also in 45 of 80 human pituitary tumors (P < 0.05). E2F1 specifically bound the hPTTG1 promoter as assessed by chromatin immunoprecipitation and biotin-streptavidin pull-down assay, indicating that hPTTG1 may act as a direct E2F1 target. Transfection of E2F1 and its partner DP1 dose-dependently activated hPTTG1 transcription up to 3-fold in p53-devoid H1299 cells but not in p53-replete HCT116 cells. E2F1 overexpression enhanced endogenous hPTTG1 mRNA and protein levels up to 3-fold in H1299 cells. The presence of endogenous p53/p21 constrained the induction, whereas knocking down either p53 or p21 in HCT116 cells restored E2F1-induced hPTTG1 transactivation and expression. Moreover, suppressing Rb by small interfering RNA concordantly elevated E2F1 and hPTTG1 protein levels. In contrast, transfection of E2F1 small interfering RNA lowered hPTTG1 levels 24 h later in HCT116 than in H1299 cells, indicating that p53 delays E2F1 action on hPTTG1. These results elucidate a mechanism for abundant tumor hPTTG1 expression, whereby Rb inactivation releases E2F1 to induce hPTTG1. This signaling pathway may underlie the requirement of PTTG1 for pituitary tumorigenesis.
Collapse
Affiliation(s)
- Cuiqi Zhou
- Department of Medicine, Cedars-Sinai Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California 90048, USA
| | | | | | | | | |
Collapse
|
39
|
Tong Y, Ben-Shlomo A, Zhou C, Wawrowsky K, Melmed S. Pituitary tumor transforming gene 1 regulates Aurora kinase A activity. Oncogene 2008; 27:6385-95. [PMID: 18663361 DOI: 10.1038/onc.2008.234] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pituitary tumor transforming gene 1 (PTTG1), a transforming gene highly expressed in several cancers, is a mammalian securin protein regulating both G1/S and G2/M phases. Using protein array screening, we showed PTTG1 interacting with Aurora kinase A (Aurora-A), and confirmed the interaction using co-immunoprecipitation, His-tagged pull-down assays and intracellular immunofluorescence colocalization. PTTG1 transfection into HCT116 cells prevented Aurora-A T288 autophosphorylation, inhibited phosphorylation of the histone H3 Aurora-A substrate and resulted in abnormally condensed chromatin. PTTG1-null cell proliferation was more sensitive to Aurora-A knock down and to Aurora kinase Inhibitor III treatment. The results indicate that PTTG1 and Aurora-A interact to regulate cellular responses to anti-neoplastic drugs. PTTG1 knockdown is therefore a potential approach to improve the efficacy of tumor Aurora kinase inhibitors.
Collapse
Affiliation(s)
- Y Tong
- Department of Medicine, Cedars-Sinai Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90048, USA
| | | | | | | | | |
Collapse
|
40
|
Chen RN, Huang YH, Yeh CT, Liao CH, Lin KH. Thyroid hormone receptors suppress pituitary tumor transforming gene 1 activity in hepatoma. Cancer Res 2008; 68:1697-706. [PMID: 18339849 DOI: 10.1158/0008-5472.can-07-5492] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pituitary tumor transforming gene 1 (PTTG1) is expressed in most tumors. However, whether thyroid hormone (T(3)) and its receptors (TR) regulate PTTG1 in human hepatocellular carcinomas (HCC) remains unclear. Previous cDNA microarrays revealed PTTG1 is down-regulated by T(3)/TR. This study investigated the significance of PTTG1 regulation by T(3) in HCC cells. The PTTG1 mRNA and protein expression were repressed by T(3) in HCC cell lines overexpressing TR. However, after knockdown of TRs expression by RNA interference, PTTG1 repression by T(3) was abolished. Similar results were observed in thyroidectomized rats. To localize the regulatory region in the PTTG1 promoter, serial deletions within the PTTG1 promoter region were constructed. The promoter activity of the PTTG1 gene was repressed (25-51%) by T(3). Additionally, these findings indicate that PTTG1 may be regulated by Sp1. The critical role of the -594 and -520 Sp1 binding sites was confirmed by electrophoretic mobility shift assay. Transfection with Sp1 expression vector enhanced the activity of the PTTG1 promoter fragment reporter. Also, Sp1 was down-regulated in HCC cells and in thyroidectomized rat after T(3) treatment. Additionally, ectopic expression of PTTG1 promotes cell proliferation in Hep3B hepatoma cells. Conversely, knockdown of PTTG1 or Sp1 expression reduced cell proliferation in HepG2 cells. Notably, the expression of PTTG1 and Sp1 was inversely correlated with the expression of TR proteins in HCC. Together, these findings indicate that PTTG1 gene expression is mediated by Sp1 and is indirectly down-regulated by T(3). Finally, overexpression of PTTG1 or SP1 in HCCs is TR-dependent and crucial in the development of HCC.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Growth Processes/genetics
- Cell Line, Tumor
- Down-Regulation/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Male
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Promoter Regions, Genetic
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Rats
- Rats, Sprague-Dawley
- Receptors, Thyroid Hormone/biosynthesis
- Receptors, Thyroid Hormone/genetics
- Receptors, Thyroid Hormone/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Securin
- Sp1 Transcription Factor/biosynthesis
- Sp1 Transcription Factor/genetics
- Sp1 Transcription Factor/metabolism
- Transfection
- Triiodothyronine/pharmacology
Collapse
Affiliation(s)
- Ruey-Nan Chen
- Department of Biochemistry, School of Medicine, Chang-Gung University, Taoyuan, Taiwan, Republic of China
| | | | | | | | | |
Collapse
|
41
|
Kim DS, Fong J, Read ML, McCabe CJ. The emerging role of pituitary tumour transforming gene (PTTG) in endocrine tumourigenesis. Mol Cell Endocrinol 2007; 278:1-6. [PMID: 17928133 DOI: 10.1016/j.mce.2007.08.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Accepted: 08/17/2007] [Indexed: 11/20/2022]
Abstract
It is now 10 years since PTTG was first cloned and isolated. Perhaps the major story of the intervening decade of work performed by numerous groups around the world is the sheer multifunctionality ascribed to this gene. PTTG has been implicated in mechanisms of gene transactivation, cell transformation, angiogenesis, metabolism, apoptosis, DNA repair, genetic instability and mitotic control, both in endocrine and non-endocrine settings. In the current review, we cast a critical eye over a decade of PTTG research within the field of endocrine neoplasia.
Collapse
Affiliation(s)
- D S Kim
- Institute of Biomedical Research, Division of Medical Sciences, University of Birmingham, Birmingham B15 2TH, UK
| | | | | | | |
Collapse
|
42
|
Boikos SA, Stratakis CA. Molecular genetics of the cAMP-dependent protein kinase pathway and of sporadic pituitary tumorigenesis. Hum Mol Genet 2007; 16 Spec No 1:R80-7. [PMID: 17613552 DOI: 10.1093/hmg/ddm019] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Pituitary tumors are among the most common human neoplasms. Although these common lesions rarely become clinically manifest and they are almost never malignant, they are the cause of significant morbidity in affected patients. The genetic causes of common pituitary tumors remain for the most part unknown; progress has been limited to the elucidation of the molecular etiology of four genetic syndromes predisposing to pituitary neoplasias: McCune-Albright syndrome, multiple endocrine neoplasia type 1, Carney complex and, most recently, familial acromegaly and prolactinomas and other tumors caused by mutations in the GNAS, menin, PRKAR1A, AIP, and p27 (CDKN1B) genes, respectively. Intense molecular studies of sporadic pituitary tumors from patients with negative family histories and no other neoplasms have yielded interesting findings with abnormalities in growth factor expression and cell cycle control dysregulation. To add to the difficulties in understanding pituitary tumorigenesis in man, good murine models of these neoplasms simply do not exist: pituitary tumors are common in rodents, but their histologic origin (mostly from the intermediate lobe), age of presentation (late in murine life) and clinical course make them hardly models of their human counterparts. The present report reviews the clinical and molecular genetics of the cAMP-dependent protein kinase pathway in human pituitary tumors; it also reviews briefly other pathways that have been involved in sporadic pituitary neoplasms. At the end, we attempt a unifying hypothesis for pituitary tumorigenesis, taking into account data that are also discussed elsewhere in this issue.
Collapse
Affiliation(s)
- Sosipatros A Boikos
- Section on Endocrinology and Genetics (SEGEN), Developmental Endocrinology Branch (DEB), National Institute of Child Health and Human Development (NICHD), National Institues of Health, Bethesda, MD 20892-1103, USA
| | | |
Collapse
|
43
|
Abstract
Pituitary tumor-transforming gene-1 (PTTG1) is overexpressed in a variety of endocrine-related tumors, especially pituitary, thyroid, breast, ovarian, and uterine tumors, as well as nonendocrine-related cancers involving the central nervous, pulmonary, and gastrointestinal systems. Forced PTTG1 expression induces cell transformation in vitro and tumor formation in nude mice. In some tumors, high PTTG1 levels correlate with invasiveness, and PTTG1 has been identified as a key signature gene associated with tumor metastasis. Increasing evidence supports a multifunctional role of PTTG1 in cell physiology and tumorigenesis. Physiological PTTG1 properties include securin activity, DNA damage/repair regulation and involvement in organ development and metabolism. Tumorigenic mechanisms for PTTG1 action involve cell transformation and aneuploidy, apoptosis, and tumorigenic microenvironment feedback. This paper reviews recent advances in our understanding of PTTG1 structure and regulation and addresses known mechanisms of PTTG1 action. Recent knowledge gained from PTTG1-null mouse models and transgenic animals and their potential application to subcellular therapeutic targeting PTTG1 are discussed.
Collapse
Affiliation(s)
- George Vlotides
- Department of Medicine, Cedars-Sinai Medical Center, University of California School of Medicine, Los Angeles, California 90048, USA
| | | | | |
Collapse
|
44
|
Cristina C, Díaz-Torga GS, Goya RG, Kakar SS, Perez-Millán MI, Passos VQ, Giannella-Neto D, Bronstein MD, Becu-Villalobos D. PTTG expression in different experimental and human prolactinomas in relation to dopaminergic control of lactotropes. Mol Cancer 2007; 6:4. [PMID: 17222350 PMCID: PMC1779802 DOI: 10.1186/1476-4598-6-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2007] [Accepted: 01/12/2007] [Indexed: 11/18/2022] Open
Abstract
Background Pituitary tumor transforming gene (pttg) is a novel oncogene that is expressed at higher level in most of the tumors analyzed to date compared to normal tissues. Nevertheless, its expression in prolactinomas and its relation with the pituitary dopamine receptor 2 (D2R) are not well defined. We sought to determine the pituitary level of pttg in three different experimental models of prolactinomas with altered dopaminergic control of the pituitary: the dopaminergic D2R knockout female mouse, the estrogen-treated rat, and the senescent female rat. These three models shared the characteristics of increased pituitary weight, hyperprolactinemia, lactotrope hyperplasia and reduced or absent dopaminergic action at the pituitary level. We also studied samples from human macroprolactinomas, which were characterized as responsive or resistant to dopamine agonist therapy. Results When compared to female wild-type mice, pituitaries from female D2R knockout mice had decreased PTTG concentration, while no difference in pttg mRNA level was found. In senescent rats no difference in pituitary PTTG protein expression was found when compared to young rats. But, in young female rats treated with a synthetic estrogen (Diethylstylbestrol, 20 mg) PTTG protein expression was enhanced (P = 0.029). Therefore, in the three experimental models of prolactinomas, pituitary size was increased and there was hyperprolactinemia, but PTTG levels followed different patterns. Patients with macroprolactinomas were divided in those in which dopaminergic therapy normalized or failed to normalize prolactin levels (responsive and resistant, respectively). When pituitary pttg mRNA level was analyzed in these macroprolactinomas, no differences were found. We next analyzed estrogen action at the pituitary by measuring pituitary estrogen receptor α levels. The D2R knockout female mice have low estrogen levels and in accordance, pituitary estrogen receptors were increased (P = 0.047). On the other hand, in senescent rats estrogen levels were slightly though not significantly higher, and estrogen receptors were similar between groups. The estrogen-treated rats had high pharmacological levels of the synthetic estrogen, and estrogen receptors were markedly lower than in controls (P < 0.0001). Finally, in patients with dopamine resistant or responsive prolactinomas no significant differences in estrogen receptor α levels were found. Therefore, pituitary PTTG was increased only if estrogen action was increased, which correlated with a decrease in pituitary estrogen receptor level. Conclusion We conclude that PTTG does not correlate with prolactin levels or tumor size in animal models of prolactinoma, and its pituitary content is not related to a decrease in dopaminergic control of the lactotrope, but may be influenced by estrogen action at the pituitary level. Therefore it is increased only in prolactinomas generated by estrogen treatment, and not in prolactinomas arising from deficient dopamine control, or in dopamine resistant compared with dopamine responsive human prolactinomas. These results are important in the search for reliable prognostic indicators for patients with pituitary adenomas which will make tumor-specific therapy possible, and help to elucidate the poorly understood phenomenon of pituitary tumorigenesis.
Collapse
Affiliation(s)
- Carolina Cristina
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas. V. Obligado 2490. (1428) Buenos Aires. Argentina
| | - Graciela S Díaz-Torga
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas. V. Obligado 2490. (1428) Buenos Aires. Argentina
| | - Rodolfo G Goya
- Institute for Biochemical Research-Histology B, Faculty of Medicine, University of La Plata, Argentina
| | - Sham S Kakar
- Department of Medicine, James Graham Brown Cancer Center. University of Louisville, Louisville, KY, USA
| | - María I Perez-Millán
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas. V. Obligado 2490. (1428) Buenos Aires. Argentina
| | - Vanessa Q Passos
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Hospital das Clinicas. University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Daniel Giannella-Neto
- Laboratory for Cellular and Molecular Endocrinology (LIM 25), Hospital das Clinicas. University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Marcello D Bronstein
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Hospital das Clinicas. University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Damasia Becu-Villalobos
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas. V. Obligado 2490. (1428) Buenos Aires. Argentina
| |
Collapse
|