Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 512
Filtrar
1.
Drug Resist Updat ; 73: 101056, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38277755

RESUMEN

BACKGROUND: The treatment of dopamine agonists (DA) resistant prolactinomas remains a formidable challenge, as the mechanism of resistance is still unclear, and there are currently no viable alternative drug therapies available. This study seeks to investigate the mechanism of DA resistance in prolactinomas and identify new potentially effective drugs. METHODS: To explore the mechanism of DA resistance in prolactinomas, this study conducted transcriptome sequencing analysis on 27 cases of DA-resistant prolactinomas and 10 cases of sensitive prolactinomas. In addition, single-cell sequencing analysis was performed on 3 cases of DA-resistant prolactinomas and 3 cases of sensitive prolactinomas. Furthermore, to screen for potential therapeutic drugs, the study successfully established an organoids model for DA-resistant prolactinomas and screened 180 small molecule compounds using 8 organoids. The efficacy of the identified drugs was verified through various assays, including CCK-8, colony formation, CTG, and flow cytometry, and their mechanisms of action were confirmed through WB and IHC. The effectiveness of the identified drugs was evaluated both in vitro and in vivo. RESULTS: The results of transcriptome sequencing and single-cell sequencing analyses showed that DA resistance in prolactinomas is associated with the upregulation of the Focal Adhesion (FA) signaling pathway. Additionally, immunohistochemical validation revealed that FAK and Paxillin were significantly upregulated in DA-resistant prolactinomas. Screening of 180 small molecule compounds using 8 organoids identified Genistein as a potentially effective drug for DA-resistant prolactinomas. Experimental validation demonstrated that Genistein inhibited the proliferation of pituitary tumor cell lines and organoids and promoted apoptosis in pituitary tumor cells. Moreover, both the cell sequencing results and WB validation results of the drug-treated cells indicated that Genistein exerts its anti-tumor effect by inhibiting the FA pathway. In vivo, experiments also showed that Genistein can inhibit subcutaneous tumor formation. CONCLUSION: DA resistance in prolactinomas is associated with upregulation of the Focal Adhesion (FA) signaling pathway, and Genistein can exert its anti-tumor effect by inhibiting the expression of the FA pathway.


Asunto(s)
Tumores Neuroendocrinos , Neoplasias Hipofisarias , Prolactinoma , Humanos , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Agonistas de Dopamina/farmacología , Agonistas de Dopamina/uso terapéutico , Prolactinoma/tratamiento farmacológico , Prolactinoma/genética , Prolactinoma/metabolismo , Prolactina/metabolismo , Prolactina/uso terapéutico , Genisteína/uso terapéutico , Tumores Neuroendocrinos/tratamiento farmacológico , Resistencia a Antineoplásicos/genética
2.
J Biochem Mol Toxicol ; 38(6): e23734, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38764151

RESUMEN

We explored the effects of curcumin on the aberrant biological behaviors of prolactinoma cells and the downstream pathways through which curcumin exerts its antitumor effects. We used quantitative reverse transcription-polymerase chain reaction assays to measure miR-206 expression levels in peripheral blood samples from patients with prolactinoma before and after curcumin treatment. We also investigated the proliferation level, viability, and invasion ability of groups of cells treated with different concentrations of curcumin using 3-(4,5)-dimethylthiahiazo (-z-y1)-3-di-phenytetrazoliumromide (MTT) assays, cell cloning assays, and Transwell assays, respectively. Furthermore, we determined the levels of autophagy-related proteins and protein kinase B/mammalian target of the rapamycin (Akt/mTOR) signaling pathway-related proteins in each group of treated cells by western blot. Curcumin treatment upregulated miR-206 expression levels in the peripheral blood of patients with prolactinoma and in GH3 cells. Knockdown of miR-206 expression enhanced the proliferation and invasive ability of GH3 cells, while curcumin treatment effectively inhibited the aberrant biological behavior of GH3 cells enhanced by miR-206 knockdown. miR-206 knockdown also activated the Akt/mTOR signaling pathway and inhibited autophagy in GH3 cells, and these changes were effectively reversed by curcumin treatment. Thus, curcumin inhibited the Akt/mTOR signaling pathway and promoted cell autophagy by miR-206 upregulation, resulting in antitumor effects that inhibited prolactinoma cell proliferation and invasion.


Asunto(s)
Autofagia , Curcumina , MicroARNs , Prolactinoma , MicroARNs/genética , MicroARNs/metabolismo , Curcumina/farmacología , Humanos , Autofagia/efectos de los fármacos , Prolactinoma/tratamiento farmacológico , Prolactinoma/patología , Prolactinoma/genética , Prolactinoma/metabolismo , Línea Celular Tumoral , Regulación hacia Arriba/efectos de los fármacos , Masculino , Serina-Treonina Quinasas TOR/metabolismo , Femenino , Neoplasias Hipofisarias/patología , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/metabolismo , Adulto , Proteínas Proto-Oncogénicas c-akt/metabolismo , Antineoplásicos/farmacología , Transducción de Señal/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Persona de Mediana Edad , Ratas
3.
J Endocrinol Invest ; 46(8): 1573-1587, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36853491

RESUMEN

PURPOSE: Prolactinomas are one of the most common pituitary neuroendocrine tumors (PitNETs), accounting for approximately 50% of all pituitary tumors. Dopamine agonists are the main treatment for prolactinoma, but a small number of patients are still resistant to pharmacotherapy. Recent discoveries have revealed that ferroptosis is involved in regulating tumor drug resistance. However, the role of ferroptosis in prolactinoma has not been reported. In this study, we aimed to explore the mechanism of a circRNA in ferroptosis in prolactinoma. METHODS: The expression of circOMA1 in prolactinoma tissues was examined by quantitative reverse transcription PCR (qRT-PCR). The biological function of circOMA1 was evaluated in vitro and in vivo. To explore the role of ferroptosis in prolactinoma, we used qRT-PCR and western blotting. Glutamate-cysteine ligase, modifier subunit (GCLM) was predicted to be a direct target gene of miR-145-5p by bioinformatics analysis, which was confirmed by luciferase reporter assays. RESULTS: circOMA1 was overexpressed in drug-resistant prolactinoma tissues compared with sensitive prolactinoma samples. We further found that circOMA1 promoted MMQ cells growth in vivo and in vitro. In addition, GCLM was directly targeted by miR-145-5p and indirectly regulated by circOMA1. Importantly, circOMA1 induced ferroptosis resistance through the increased expression of Nrf2, GPX4, and xCT, and circOMA1 attenuated CAB-induced ferroptosis in MMQ cells in vivo and in vitro. CONCLUSION: The present study demonstrates that circOMA1 attenuates CAB efficacy through ferroptosis resistance and may be a new therapeutic target for the individualized treatment of DA-resistant prolactinoma patients.


Asunto(s)
Ferroptosis , MicroARNs , Neoplasias Hipofisarias , Prolactinoma , Humanos , Prolactinoma/tratamiento farmacológico , Prolactinoma/genética , Prolactinoma/metabolismo , Cabergolina/uso terapéutico , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Agonistas de Dopamina/uso terapéutico , MicroARNs/genética , MicroARNs/uso terapéutico
4.
JAMA ; 329(16): 1386-1398, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37097352

RESUMEN

Importance: Pituitary adenomas are neoplasms of the pituitary adenohypophyseal cell lineage and include functioning tumors, characterized by the secretion of pituitary hormones, and nonfunctioning tumors. Clinically evident pituitary adenomas occur in approximately 1 in 1100 persons. Observations: Pituitary adenomas are classified as either macroadenomas (≥10 mm) (48% of tumors) or microadenomas (<10 mm). Macroadenomas may cause mass effect, such as visual field defects, headache, and/or hypopituitarism, which occur in about 18% to 78%, 17% to 75%, and 34% to 89% of patients, respectively. Thirty percent of pituitary adenomas are nonfunctioning adenomas, which do not produce hormones. Functioning tumors are those that produce an excess of normally produced hormones and include prolactinomas, somatotropinomas, corticotropinomas, and thyrotropinomas, which produce prolactin, growth hormone, corticotropin, and thyrotropin, respectively. Approximately 53% of pituitary adenomas are prolactinomas, which can cause hypogonadism, infertility, and/or galactorrhea. Twelve percent are somatotropinomas, which cause acromegaly in adults and gigantism in children, and 4% are corticotropinomas, which secrete corticotropin autonomously, resulting in hypercortisolemia and Cushing disease. All patients with pituitary tumors require endocrine evaluation for hormone hypersecretion. Patients with macroadenomas additionally require evaluation for hypopituitarism, and patients with tumors compressing the optic chiasm should be referred to an ophthalmologist for formal visual field testing. For those requiring treatment, first-line therapy is usually transsphenoidal pituitary surgery, except for prolactinomas, for which medical therapy, either bromocriptine or cabergoline, is usually first line. Conclusions and Relevance: Clinically manifest pituitary adenomas affect approximately 1 in 1100 people and can be complicated by syndromes of hormone excess as well as visual field defects and hypopituitarism from mass effect in larger tumors. First-line therapy for prolactinomas consists of bromocriptine or cabergoline, and transsphenoidal pituitary surgery is first-line therapy for other pituitary adenomas requiring treatment.


Asunto(s)
Adenoma , Neoplasias Hipofisarias , Adulto , Niño , Femenino , Humanos , Embarazo , Adenoma/complicaciones , Adenoma/diagnóstico , Adenoma/metabolismo , Adenoma/terapia , Hormona Adrenocorticotrópica/biosíntesis , Bromocriptina/uso terapéutico , Cabergolina/uso terapéutico , Hormona de Crecimiento Humana/biosíntesis , Hipopituitarismo/diagnóstico , Hipopituitarismo/etiología , Hipopituitarismo/metabolismo , Hipopituitarismo/terapia , Neoplasias Hipofisarias/complicaciones , Neoplasias Hipofisarias/diagnóstico , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/terapia , Prolactinoma/diagnóstico , Prolactinoma/etiología , Prolactinoma/metabolismo , Prolactinoma/terapia
5.
Tumour Biol ; 44(1): 85-105, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35811548

RESUMEN

INTRODUCTION: Prolactinomas are the most frequent pituitary tumor subtype. Despite most of them respond to medical treatment, a proportion are resistant and become a challenge in clinical management. Wnt/ß-Catenin pathway has been implicated in several cancers including pituitary tumors and other sellar region malignancies. Interestingly, Wnt/ß-Catenin inhibition augments the cytotoxicity of the chemotherapeutic agent Temozolomide (TMZ) in different cancers. TMZ is now being implemented as rescue therapy for aggressive pituitary adenoma treatment. However, the molecular mechanisms associated with TMZ action in pituitary tumors remain unclear. OBJECTIVES: Our aims in the present study were to evaluate differential ß-Catenin expression in human resistant prolactinomas and Wnt/ß-Catenin signaling activation and involvement in Prolactin (PRL) secreting experimental models treated with TMZ. RESULTS: We first evaluated by immunohistochemistry ß-Catenin localization in human resistant prolactinomas in which we demonstrated reduced membrane ß-Catenin in prolactinoma cells compared to normal pituitaries, independently of the Ki-67 proliferation indexes. In turn, in vivo 15 mg/kg of orally administered TMZ markedly reduced PRL production and increased prolactinoma cell apoptosis in mice bearing xenografted prolactinomas. Intratumoral ß-Catenin strongly correlated with Prl and Cyclin D1, and importantly, TMZ downregulated both ß-Catenin and Cyclin D1, supporting their significance in prolactinoma growth and as candidates of therapeutic targets. When tested in vitro, TMZ directly reduced MMQ cell viability, increased apoptosis and produced G2/M cell cycle arrest. Remarkably, ß-Catenin activation and VEGF secretion were inhibited by TMZ in vitro. CONCLUSIONS: We concluded that dopamine resistant prolactinomas undergo a ß-Catenin relocalization in relation to normal pituitaries and that TMZ restrains experimental prolactinoma tumorigenicity by reducing PRL production and ß-Catenin activation. Together, our findings contribute to the understanding of Wnt/ß-Catenin implication in prolactinoma maintenance and TMZ therapy, opening the opportunity of new treatment strategies for aggressive and resistant pituitary tumors.


Asunto(s)
Neoplasias Hipofisarias , Prolactinoma , Animales , Ciclina D1 , Humanos , Ratones , Modelos Teóricos , Neoplasias Hipofisarias/patología , Prolactina/metabolismo , Prolactina/uso terapéutico , Prolactinoma/tratamiento farmacológico , Prolactinoma/metabolismo , Prolactinoma/patología , Temozolomida/farmacología , Temozolomida/uso terapéutico , beta Catenina
6.
Endocr Regul ; 56(4): 279-283, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36270341

RESUMEN

Objective. Prolactinoma, as a common endocrine disorder and the most frequent type of pituitary tumor, acts primarily as a suppressor on the gonadal functions. It is generally successfully treated with dopamine agonists; however, treatment resistance still remains in an unneglectable ratio. In this study, we aimed to identify factors, which may play a role in the treatment response. Methods. Seventy-six patients with prolactinoma, who have been routinely followed between 2018 and 2022 in Istanbul Research and Educational Hospital Endocrinology Outpatient Clinic, were included into the study. Initial prolactin level, adenoma size, baseline weight, body mass index (BMI), glucose, high-density lipoprotein (HDL) cholesterol, low-density lipoprotein (LDL) cholesterol, and triglyceride levels were obtained from the patient's medical records. The patients were divided into two groups: treatment respondent and non-respondent (refractory) ones, according to treatment response in the duration as suggested by the guidelines. The treatment respondent and non-respondent groups were compared according to the initial and the 3rd month prolactin levels, adenoma size, weight, BMI, and metabolic values. Results. The initial tumor diameter was 15.27±10.62 mm in the refractory and 7.42±4.42 mm in the treatment respondent groups (p=0.01). The refractory group had higher prolactin baseline level 269.96±275.78 µg/l vs. 124.55±67.35 µg/l of the respondent group (p=0.01). The refractory group had higher the 3rd month prolactin level 50.97±52.55 µg/l vs. 29.70±27.31 µg/l of the respondent group (p=0.04). The refractory group had higher frequency of cystic/hemorrhagic adenoma (47.6%, n=11/21) (p=0.01), baseline pituitary failure (33.3%, n=7/21) (p=0.01), and baseline cavernous sinus invasion (25.8, n=5/21) (p=0.01). The treatment respondent group had lower initial body weight (69.54±17.51 kg vs. 83.29±16.21 kg) (p<0.01), and lower BMI (25.98±5.47 kg/m2 vs. 27.69±6.42 kg/m2) (p=0.02). Conclusions. In this study, initial tumor size, male gender, weight, BMI, the 3rd month prolactin level, initial pituitary deficiency, and cystic/hemorrhagic component in pituitary imaging in patients with prolactinoma were associated with a lower treatment response.


Asunto(s)
Adenoma , Neoplasias Hipofisarias , Prolactinoma , Humanos , Masculino , Prolactinoma/diagnóstico por imagen , Prolactinoma/tratamiento farmacológico , Prolactinoma/metabolismo , Cabergolina/uso terapéutico , Agonistas de Dopamina/uso terapéutico , Prolactina/uso terapéutico , Ergolinas/uso terapéutico , Neoplasias Hipofisarias/diagnóstico por imagen , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/metabolismo , Adenoma/tratamiento farmacológico , Peso Corporal , Glucosa , Lipoproteínas LDL , Lipoproteínas HDL , Triglicéridos
7.
Hum Mol Genet ; 28(6): 1023-1037, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30445560

RESUMEN

Prolactinomas are the most frequent type of pituitary tumors, which represent 10-20% of all intracranial neoplasms in humans. Prolactinomas develop in mice lacking the prolactin receptor (PRLR), which is a member of the cytokine receptor superfamily that signals via Janus kinase-2-signal transducer and activator of transcription-5 (JAK2-STAT5) or phosphoinositide 3-kinase-Akt (PI3K-Akt) pathways to mediate changes in transcription, differentiation and proliferation. To elucidate the role of the PRLR gene in human prolactinomas, we determined the PRLR sequence in 50 DNA samples (35 leucocytes, 15 tumors) from 46 prolactinoma patients (59% males, 41% females). This identified six germline PRLR variants, which comprised four rare variants (Gly57Ser, Glu376Gln, Arg453Trp and Asn492Ile) and two low-frequency variants (Ile76Val, Ile146Leu), but no somatic variants. The rare variants, Glu376Gln and Asn492Ile, which were in complete linkage disequilibrium, and are located in the PRLR intracellular domain, occurred with significantly higher frequencies (P < 0.0001) in prolactinoma patients than in 60 706 individuals of the Exome Aggregation Consortium cohort and 7045 individuals of the Oxford Biobank. In vitro analysis of the PRLR variants demonstrated that the Asn492Ile variant, but not Glu376Gln, when compared to wild-type (WT) PRLR, increased prolactin-induced pAkt signaling (>1.3-fold, P < 0.02) and proliferation (1.4-fold, P < 0.02), but did not affect pSTAT5 signaling. Treatment of cells with an Akt1/2 inhibitor or everolimus, which acts on the Akt pathway, reduced Asn492Ile signaling and proliferation to WT levels. Thus, our results identify an association between a gain-of-function PRLR variant and prolactinomas and reveal a new etiology and potential therapeutic approach for these neoplasms.


Asunto(s)
Susceptibilidad a Enfermedades , Prolactinoma/etiología , Prolactinoma/metabolismo , Receptores de Prolactina/genética , Receptores de Prolactina/metabolismo , Alelos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Everolimus/farmacología , Femenino , Genotipo , Humanos , Quinasas Janus/metabolismo , Masculino , Mutación , Prolactinoma/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Prolactina/química , Factores de Transcripción STAT/metabolismo , Transducción de Señal
8.
Horm Metab Res ; 53(7): 413-424, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34282593

RESUMEN

Despite most of the prolactinomas can be treated with endocrine therapy and/or surgery, a significant percentage of these tumors can be resistant to endocrine treatments and/or recur with prominent invasion into the surrounding anatomical structures. Hence, clinical, pathological, and molecular definitions of aggressive prolactinomas are important to guide for classical and novel treatment modalities. In this review, we aimed to define molecular endocrinological features of dopamine agonist-resistant and aggressive prolactinomas for designing future multimodality treatments. Besides surgery, temozolomide chemotherapy and radiotherapy, peptide receptor radionuclide therapy, estrogen pathway modulators, progesterone antagonists or agonists, mTOR/akt inhibitors, pasireotide, gefitinib/lapatinib, everolimus, and metformin are tested in preclinical models, anecdotal cases, and in small case series. Moreover, chorionic gonadotropin, gonadotropin releasing hormone, TGFß and PRDM2 may seem like possible future targets for managing aggressive prolactinomas. Lastly, we discussed our management of a unique prolactinoma case by asking which tumors' proliferative index (Ki67) increased from 5-6% to 26% in two subsequent surgeries performed in a 2-year period, exerted massive invasive growth, and secreted huge levels of prolactin leading up to levels of 1 605 671 ng/dl in blood.


Asunto(s)
Agonistas de Dopamina/farmacología , Resistencia a Antineoplásicos , Prolactinoma/terapia , Terapia Combinada , Humanos , Pronóstico , Prolactinoma/metabolismo , Prolactinoma/patología
9.
BMC Endocr Disord ; 21(1): 81, 2021 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-33902531

RESUMEN

BACKGROUND: Hyperprolactinaemia might cause adverse metabolic effects. The aim of our study was to compare parameters of body composition, glucose and lipid metabolism between untreated patients with prolactinoma and controls and to assess changes after initiation of cabergoline. METHODS: Case-control study with a retrospectively analyzed follow-up in patients with prolactinoma after initiation of cabergoline therapy. RESULTS: 21 patients with prolactinoma (9 micro- and 12 macroprolactinomas; 7 females) and 30 controls were analyzed. Patients with prolactinoma had significantly higher BMI than controls; fat mass did not differ between groups. Only men - but not women - with prolactinoma had significantly higher fat mass at all six sites measured compared to controls. Levels of LDL (130 (107-147.5) vs. 94.5 (80-127.5) mg/dl, p < 0.001) were significantly higher, levels of HDL (56 ± 16.7 vs. 69.2 ± 14.6 mg/dl, p = 0.004) significantly lower than in controls. Fasting glucose, HOMA-IR, HbA1c, adiponectin, CRP, and homocysteine did not differ between groups. After a median of 10 weeks (IQR 7-18 weeks) after initiation of cabergoline, total (from 212.5 ± 36.2 to 196.9 ± 40.6 mg/dl, p = 0.018) and LDL cholesterol (130 (107-147.5) to 106.5 (94.3-148) mg/dl, p = 0.018) had significantly decreased. Analyzing men and women separately, this change occurred in men only. CONCLUSIONS: Reasons for the association between prolactin and metabolic parameters include direct effects of prolactin on adipose tissue, hyperprolactinaemia-triggered hypogonadism and dopamine-agonist therapy per se. Altered lipid metabolism in patients with prolactinoma might imply an increased cardiovascular risk, highlighting the necessity to monitor metabolic parameters in these patients.


Asunto(s)
Composición Corporal , Hiperprolactinemia/metabolismo , Metabolismo de los Lípidos , Neoplasias Hipofisarias/metabolismo , Prolactinoma/metabolismo , Adiposidad/efectos de los fármacos , Adiposidad/fisiología , Adulto , Austria , Composición Corporal/efectos de los fármacos , Índice de Masa Corporal , Cabergolina/uso terapéutico , Estudios de Casos y Controles , Dislipidemias/tratamiento farmacológico , Dislipidemias/etiología , Dislipidemias/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Hiperprolactinemia/complicaciones , Hiperprolactinemia/tratamiento farmacológico , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Sobrepeso/tratamiento farmacológico , Sobrepeso/etiología , Sobrepeso/metabolismo , Neoplasias Hipofisarias/complicaciones , Neoplasias Hipofisarias/tratamiento farmacológico , Prolactinoma/complicaciones , Prolactinoma/tratamiento farmacológico , Estudios Retrospectivos , Adulto Joven
10.
Pituitary ; 24(1): 79-95, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33025546

RESUMEN

PURPOSE: Prolactinomas are the most prevalent functioning pituitary adenomas. They affect gonadal function as well as health-related quality of life (HRQoL). This study aimed to report healthcare utilization and costs, including their determinants, for prolactinoma patients. METHODS: Cross-sectional study of 116 adult prolactinoma patients in chronic care in a Dutch tertiary referral center. Patients completed four validated questionnaires, assessing healthcare utilization and costs over the previous 12 months (Medical Consumption Questionnaire), disease bother and needs (Leiden Bother and Needs Questionnaire Pituitary), HRQoL (Short Form-36), and self-reported health status (EuroQol 5D). Regression analyses were used to assess associations between disease-related characteristics and healthcare utilization and costs. RESULTS: Mean age was 52.0 years (SD 13.7) and median follow-up was 15.0 years (IQR 7.6-26.1). Patients visited the endocrinologist (86.2%), general practitioner (37.9%), and ophthalmologist (25.0%) most frequently. Psychological care was used by 12.9% of patients and 5% were admitted to hospital. Mean annual healthcare costs were €1928 (SD 3319), mainly for pituitary-specific medication (37.6% of total costs), hospitalization (19.4%) and specialist care (16.1%). Determinants for higher healthcare utilization and costs were greater disease bother and needs for support, lower HRQoL, elevated prolactin, and longer disease duration, while tumor size, hypopituitarism and adrenal insufficiency were not significantly associated with healthcare utilization and costs. CONCLUSION: Healthcare utilization and costs of prolactinoma patients are related to patient-reported HRQoL, bother by disease and needs for support. Therefore, addressing patients' HRQoL and needs is a way forward to improve efficiency of care and patients' health status.


Asunto(s)
Prolactinoma/metabolismo , Adulto , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Prolactinoma/genética , Calidad de Vida , Análisis de Regresión , Encuestas y Cuestionarios
11.
Pituitary ; 24(1): 48-52, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33010004

RESUMEN

Pathogenic variants in the aryl hydrocarbon receptor-interacting protein (AIP) gene are increasingly recognised as a cause of familial isolated pituitary adenoma. AIP-associated tumours are most commonly growth hormone (GH) producing. In our cohort of 175 AIP mutation positive patients representing 93 kindreds, 139 (79%) have GH excess, 19 have prolactinoma (17 familial and 2 sporadic cases) and out of the 17 clinically non-functioning tumours 4 were subsequently operated and found to be GH or GH & prolactin immunopositive adenoma. Here we report a family with an AIP variant, in which multiple family members are affected by prolactinoma, but none with GH excess. To our knowledge this is the first reported family with an AIP pathogenic variant to be affected solely by prolactinoma. These data suggest that prolactinoma families represent a small subset of AIP mutation positive kindreds, and similar to young-onset sporadic prolactinomas, AIP screening would be indicated.


Asunto(s)
Adenoma Hipofisario Secretor de Hormona del Crecimiento/epidemiología , Prolactinoma/epidemiología , Adenoma/epidemiología , Adenoma/metabolismo , Adulto , Femenino , Pruebas Genéticas , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Linaje , Neoplasias Hipofisarias/epidemiología , Neoplasias Hipofisarias/metabolismo , Prolactinoma/metabolismo
12.
Int J Mol Sci ; 22(20)2021 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-34681905

RESUMEN

Prolactinoma has the highest incidence rate among patients with functional pituitary tumours. Although mostly benign, there is a subgroup that can be aggressive. Some clinical, radiological and pathology features have been associated with a poor prognostic. Therefore, it can be considered as a group of heterogeneous tumours. The aim of this paper is to give an overview of the molecular pathways involved in the behaviour of prolactinoma in order to improve our approach and gain deeper insight into the better understanding of tumour development and its management. This is essential for identifying patients harbouring aggressive prolactinoma and to establish personalised therapeutics options.


Asunto(s)
Antineoplásicos/uso terapéutico , Agonistas de Dopamina/uso terapéutico , Neoplasias Hipofisarias/tratamiento farmacológico , Prolactinoma/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Humanos , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Prolactinoma/metabolismo , Prolactinoma/patología
13.
Ter Arkh ; 93(10): 1234-1239, 2021 Oct 15.
Artículo en Ruso | MEDLINE | ID: mdl-36286827

RESUMEN

The prevalence of hyperprolactinemia in postmenopausal women is unknown and has been estimated as infrequent by many studies. Prolactinomas found after menopause are usually macroadenomas and remain unrecognized for a long time due to atypical clinical signs or their absence. The growth potential of prolactinomas persists after menopause, most of them are invasive and accompanied by high prolactin levels. Treatment with dopamine agonists is usually long-term, the goals of which are to reduce tumor size, normalize prolactin levels and the negative effects of hyperprolactinemia. Treatment with cabergoline makes it possible to achieve remission of the disease in the first years after discontinuation, however, the proportion of relapses in postmenopausal women increases 5 years after discontinuation of the drug. Remission of prolactinomas is not evident in postmenopausal women. The modern management of patients with prolactinoma and/or hyperprolactinemia does not have clear positions in the postmenopausal period. Controversial issues remain: an ambiguous relationship between prolactin levels and breast cancer, there are no convincing conclusions on the improvement of bone mineral density and/or a decrease in the risk of fractures with normalization of prolactin levels, there are no data on metabolic parameters after the end of treatment with dopamine agonists, conflicting information about the relationship of prolactin levels and the severity of the manifold manifestations of the climacteric syndrome. The use of estrogen-progestin drugs in women with hyperprolactinemia/prolactinomas is also not well understood. Thus, the problem of hyperprolactinemia in the perimenopausal and postmenopausal period is underestimated and requires additional research, as well as the development of diagnostic and therapeutic strategies for potential benefits in terms of weight loss, improving insulin sensitivity, reducing the risk of fractures, maintaining sexuality and psycho-emotional well-being.


Asunto(s)
Hiperprolactinemia , Neoplasias Hipofisarias , Prolactinoma , Humanos , Femenino , Hiperprolactinemia/diagnóstico , Hiperprolactinemia/tratamiento farmacológico , Hiperprolactinemia/metabolismo , Prolactinoma/diagnóstico , Prolactinoma/tratamiento farmacológico , Prolactinoma/metabolismo , Cabergolina/uso terapéutico , Posmenopausia , Agonistas de Dopamina/uso terapéutico , Prolactina/metabolismo , Prolactina/uso terapéutico , Progestinas/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Estrógenos
14.
Acta Neuropathol ; 140(3): 377-396, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32572597

RESUMEN

Pituitary adenoma (PA) is one of the most common intracranial tumors, and approximately 40% of all PAs are prolactinomas. Dopamine agonists (DAs), such as cabergoline (CAB), have been successfully used in the treatment of prolactinomas. The expression of dopamine type 2 receptor (DRD2) determines the therapeutic effect of DAs, but the molecular mechanisms of DRD2 regulation are not fully understood. In this study, we first demonstrated that DRD2 underwent proteasome-mediated degradation. We further employed the yeast two-hybrid system and identified kelch repeat and BTB (POZ) domain containing 7 (KBTBD7), a substrate adaptor for the CUL3-RING ubiquitin (Ub) ligase complex, as a DRD2-interacting protein. KBTBD6/7 directly interacted with, and ubiquitinated DRD2 at five ubiquitination sites (K221, K226, K241, K251, and K258). CAB, a high-affinity DRD2 agonist, induced DRD2 internalization, and cytoplasmic DRD2 was degraded via ubiquitination under the control of KBTBD6/7, the activity of which attenuated CAB-mediated inhibition of the AKT/mTOR pathway. KBTBD7 knockout (KO) mice were generated using the CRISPR-Cas9 technique, in which the static level of DRD2 protein was elevated in the pituitary gland, thalamus, and heart, compared to that of WT mice. Consistently, the expression of KBTBD6/7 was negatively correlated with that of DRD2 in human pituitary tumors. Moreover, KBTBD7 was highly expressed in dopamine-resistant prolactinomas, but at low levels in dopamine-sensitive prolactinomas. Knockdown of KBTBD6/7 sensitized MMQ cells and primary pituitary tumor cells to CAB treatment. Conversely, KBTBD7 overexpression increased CAB resistance of estrogen-induced in situ rat prolactinoma model. Together, our findings have uncovered the novel mechanism of DRD2 protein degradation and shown that the KBTBD6/7-DRD2 axis regulates PA sensitivity to DA treatment. KBTBD6/7 may thus become a promising therapeutic target for pituitary tumors.


Asunto(s)
Adenoma/tratamiento farmacológico , Agonistas de Dopamina/uso terapéutico , Hipófisis/efectos de los fármacos , Neoplasias Hipofisarias/tratamiento farmacológico , Adenoma/metabolismo , Animales , Dopamina/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones Noqueados , Hipófisis/patología , Neoplasias Hipofisarias/metabolismo , Prolactinoma/tratamiento farmacológico , Prolactinoma/metabolismo , Prolactinoma/patología , Receptores de Dopamina D2/efectos de los fármacos , Receptores de Dopamina D2/metabolismo
15.
BMC Endocr Disord ; 20(1): 138, 2020 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-32894113

RESUMEN

BACKGROUND: The specific underlying pathogenesis of prolactinoma has not been clarified yet, to the best of our knowledge. p38 mitogen-activated protein kinase (MAPK) signaling including p38α MAPK (MAPK14), p38ß (MAPK11), p38γ (MAPK12) and p38δ (MAPK13) is associated with the development and progression of several types of cancer. METHODS: Immunofluorescence analysis was performed on the prolactin (PRL) and MAPK14 expressions of pituitary gland in C57BL/6 mice and human prolactinoma specimen. In the present study, the role of MAPK14 in prolactinoma was determined using estradiol-induced mice and dopamine D2 receptor knockout (DRD2-/-) mice models in C57BL/6 wild-type (WT), MAPK14-/- and DRD2-/-MAPK14+/- mice. GH3 cells were transfected with different sets of MAPK14 small interfering RNA, which to study MAPK14 and PRL expression in GH3 cells. RESULTS: Immunofluorescence analysis showed that PRL and MAPK14 expression were colocalized and increased in the pituitary gland of mice and human prolactinoma specimen compared with the control specimen. It was shown that PRL and MAPK14 expression was colocalized and increased significantly in the pituitary gland of estradiol-injected prolactinoma mice compared with the control mice. Knockout of MAPK14 significantly inhibited tumor overgrowth, and PRL expression was decreased in estradiol-induced mice. Furthermore, MAPK14 knockout of DRD2-/-MAPK14+/- mice significantly reduced the overgrowth of pituitary gland and PRL production and secretion compared with DRD2-/- mice. MAPK14 knockout using siRNA inhibited PRL production in GH3 cells. CONCLUSION: These results suggest that MAPK14 serves a promoting role in the formation of prolactinoma, and highlights the potential of MAPK14 as a potential therapeutic target in the treatment of prolactinoma.


Asunto(s)
Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Neoplasias Hipofisarias/patología , Prolactinoma/patología , ARN Interferente Pequeño/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Femenino , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 14 Activada por Mitógenos/genética , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Prolactina/genética , Prolactina/metabolismo , Prolactinoma/genética , Prolactinoma/metabolismo , Ratas , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Somatotrofos/metabolismo , Somatotrofos/patología
16.
Pituitary ; 23(1): 9-15, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31873848

RESUMEN

Prolactinomas are the most frequently seen pituitary adenomas in clinical practice. A correct biochemical diagnosis of hyperprolactinemia is a prerequisite for further investigation but may be hampered by analytical difficulties as well as a large number of potentially overlapping conditions associated with increased prolactin levels. Suspicion should rise in patients whose symptoms and biochemical results do not match. Assay problems, macroprolactinemia, and high-dose hook effect are discussed as possible reasons for false positive or false negative prolactin levels. Physiological and pathological causes of hyperprolactinemia and their implications for interpreting prolactin results are reviewed.


Asunto(s)
Prolactinoma/diagnóstico , Humanos , Hiperprolactinemia/diagnóstico , Hiperprolactinemia/metabolismo , Prolactina/metabolismo , Prolactinoma/metabolismo
17.
Pituitary ; 23(6): 622-629, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32715387

RESUMEN

PURPOSE: Patients with sellar masses (SM) frequently have secondary hormonal deficiency (SHD) at initial presentation. While larger SM are more likely to present with SHD, it is unclear whether SHD at presentation is influenced by the type of SM. METHODS: We conducted a retrospective analysis of patients with SM prospectively enrolled in our comprehensive provincial neuropituitary registry between November 2005 and December 2018. SM were subdivided based on size: < 1 cm, 1-1.9 cm, 2-2.9 cm, and > 3 cm. RESULTS: A total of 914 patients met the inclusion criteria, including: 346 nonfunctioning adenomas (NFA), 261 prolactinomas (PRLoma), 51 growth hormone adenomas, 36 adrenocorticotropic adenomas, 93 Rathke's cleft cysts, 70 craniopharyngiomas and 57 meningiomas. The overall rate of SHD at presentation was highest in PRLoma (62.8%) and craniopharyngiomas (64.3%) and lowest in meningiomas (14%). While larger SM were significantly more likely to have SHD, the rate of SHD within each group was significantly different despite similar size (p < 0.001). Of the two largest groups of SM (NFA and PRLoma), NFA had significantly higher odds ratio (3.34, CI 1.89-5.89) of having multiple SHDs when compare with PRLoma, even when corrected for age, gender and size of tumor (p < 0.001). CONCLUSION: Our study shows that the rate and distribution of SHD in SM vary dependent upon the size of the tumor and specific pathology; in particular, NFA are more likely to present with multiple SHDs. Our data will help clinicians in determining adequate hormonal testing strategy for different SM.


Asunto(s)
Adenoma/metabolismo , Prolactinoma/metabolismo , Adulto , Craneofaringioma/metabolismo , Femenino , Humanos , Masculino , Meningioma/metabolismo , Persona de Mediana Edad , Estudios Retrospectivos
18.
Pituitary ; 23(3): 232-245, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32026205

RESUMEN

PURPOSE: The regulatory effects of estradiol on pituitary homeostasis have been well documented. However, the expression patterns of ERα66 and ERα36 and their correlations with the clinical course of postoperative prolactinoma tumors remain unclear. METHODS: The expression of ERα36, ERα66, Ki67, p53, and CD31 were determined by immunohistochemistry in 62 prolactinoma patients. Snap-frozen tumors and normal pituitaries were also examined by western blotting for estrogen receptor detection. RESULTS: A broad expression of ERα36 was identified in normal pituitaries. The median scores of ERα36 and ERα66 expression were 8 and 6 in normal pituitaries and 4 and 0 in tumors, respectively. Four phenotypes of ERα36 and ERα66 expression were explored in tumors with regard to sex, invasiveness, dopamine resistance, and recurrence. Low ERα36 expression was associated with tumor invasion and increased Ki67. Low ERα66 expression was associated with tumor invasion, dopamine-agonist resistance, and enhanced tumor size. Multivariable logistic regression analysis showed that low ERα36 expression is an independent risk factor for invasiveness. The significant inverse association of ERα66 with invasiveness, dopamine resistance, and tumor size remained significant after adjustment for sex as a potential confounder. After controlling for sex, the low ERα66/low ERα36 phenotype was 6.24 times more prevalent in invasive tumors than in noninvasive tumors. Although the decreasing trend of CD31 expression from surrounding nontumoral lactotroph adenomas to tumors was similar to that of the estrogen receptors, a significant correlation was not observed here. CONCLUSION: The decreasing trends of ERα36 and ERα66 expression from normal pituitaries to tumors are associated with aggressive clinical behavior.


Asunto(s)
Biomarcadores/metabolismo , Receptor alfa de Estrógeno/metabolismo , Neoplasias Hipofisarias/metabolismo , Prolactinoma/metabolismo , Isoformas de Proteínas/metabolismo , Adulto , Western Blotting , Receptor alfa de Estrógeno/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias Hipofisarias/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Prolactinoma/genética , Isoformas de Proteínas/genética
19.
Med Sci Monit ; 26: e924884, 2020 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-32964894

RESUMEN

BACKGROUND The aim of this study was to review outcomes of gamma knife radiosurgery (GKRS) for prolactinoma and report our experience with it. MATERIAL AND METHODS We reviewed the patient database in our center and identified 24 patients with prolactinoma who underwent GKRS from 1993 to 2016.  Complete endocrine, clinical, and radiological data were available on these individuals before and after GKRS. RESULTS Data from 5 males and 19 females with a median age of 30.5 years (range, 18.1 to 51.1) were reviewed. The median follow-up was 109.3 months (range, 23.2-269.3). The median margin dose of GKRS was 15 Gy (range, 10.5 to 23.6). In total, prolactin (PRL) normalization after GKRS was achieved in 66.7% of patients. Endocrine remission (normal PRL levels after discontinuation of dopamine agonists) was achieved in 10 patients (41.7%), and endocrine control (normal PRL levels while taking dopamine agonists) was achieved in 6 patients (25.0%). All of the patients showed tumor control. New-onset hypopituitarism post-GKRS occurred in 4 patients (16.7%). No new visual dysfunction or cranial nerve dysfunction were observed after GKRS. CONCLUSIONS For treatment of prolactinomas, GKRS may provide relatively high rates of endocrine remission and tumor control, as well as a low rate of new-onset hypopituitarism. GKRS may be an effective and safe treatment for prolactinomas.


Asunto(s)
Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/radioterapia , Prolactinoma/metabolismo , Prolactinoma/radioterapia , Radiocirugia , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Resultado del Tratamiento
20.
Int J Mol Sci ; 21(6)2020 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-32188093

RESUMEN

Estrogen signaling plays an important role in pituitary development and function. In sensitive rat or mice strains of both sexes, estrogen treatments promote lactotropic cell proliferation and induce the formation of pituitary adenomas (dominantly prolactin or growth-hormone-secreting ones). In male patients receiving estrogen, treatment does not necessarily result in pituitary hyperplasia, hyperprolactinemia or adenoma development. In this review, we comprehensively analyze the mechanisms of estrogen action upon their application in male animal models comparing it with available data in human subjects. Sex-specific molecular targets of estrogen action in lactotropic (PRL) cells are highlighted in the context of their proliferative and secretory activity. In addition, putative effects of estradiol on the cellular/tumor microenvironment and the contribution of postnatal pituitary progenitor/stem cells and transdifferentiation processes to prolactinoma development have been analyzed. Finally, estrogen-induced morphological and hormone-secreting changes in pituitary thyrotropic (TSH) and adrenocorticotropic (ACTH) cells are discussed, as well as the putative role of the thyroid and/or glucocorticoid hormones in prolactinoma development, based on the current scarce literature.


Asunto(s)
Estrógenos/efectos adversos , Hiperplasia/metabolismo , Enfermedades de la Hipófisis/metabolismo , Prolactinoma/metabolismo , Adenoma/patología , Hormona Adrenocorticotrópica/metabolismo , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Estradiol , Femenino , Humanos , Hiperplasia/patología , Masculino , Ratones , Enfermedades de la Hipófisis/patología , Hipófisis/metabolismo , Neoplasias Hipofisarias/patología , Prolactina , Prolactinoma/patología , Ratas , Células Madre , Tirotropina/metabolismo , Microambiente Tumoral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA